Open access peer-reviewed chapter

Coumarin Derivatives with Antimicrobial and Antioxidant Activities

Written By

Gabriela Tataringa and Ana Maria Zbancioc

Submitted: 04 May 2019 Reviewed: 17 June 2019 Published: 27 July 2019

DOI: 10.5772/intechopen.88096

From the Edited Volume

Phytochemicals in Human Health

Edited by Venketeshwer Rao, Dennis Mans and Leticia Rao

Chapter metrics overview

1,425 Chapter Downloads

View Full Metrics

Abstract

Coumarin derivatives are structurally interesting compounds for synthesizing antimicrobial and antioxidant agents. Starting from 4-methyl-7-hydroxycoumarin, several derivatives with these properties have been obtained through different reaction steps. Their molecular structures were established by Fourier-transform infrared spectroscopy and nuclear magnetic resonance spectroscopy. The synthesized coumarin derivatives exerted meaningful activities against Gram-positive and Gram-negative bacteria as well as strains of Candida spp. All compounds also exhibited high and moderate antioxidant activity in assays for DPPH inhibition, total reducing power, and nitric oxide (NO) inhibition when compared to ascorbic acid.

Keywords

  • 4-methyl-7-hydroxycoumarin
  • synthesis
  • coumarin derivatives
  • antibacterial activity
  • antifungal activity
  • antioxidant activity

1. Introduction

Natural and synthetic coumarins have drawn much attention due to its broad pharmacological activities. Literature review reveals that coumarin (2-oxo-2H-chromene) and its derivatives represent one of the most active classes of heterocyclic compounds which possess a wide spectrum of biological activities [1, 2, 3, 4, 5, 6, 7, 8, 9]: antitumor [1, 2], antibacterial [3, 4], antifungal [5, 6, 7], anticoagulant [8], antioxidant [9], and anti-inflammatory [10].

1.1 Coumarins as antimicrobial agents

Over the past few decades, the search for newer antimicrobials remains an area of intensive investigation in the field of medicinal chemistry due to resistance developed by microorganism to conventional antibiotics. Antimicrobials are one of most significant weapons in fighting bacterial infections. Throughout history, there has been a continual battle between humans and the multitude of microorganisms that cause infection and disease [11, 12]. Coumarin derivatives have a wide range of structural modifications [13], and they can serve as molecular templates for new drugs. Coumarin derivatives are also considered as potential antimicrobial agents [14].

Medimagh-Saidana et al. reported synthesis and antimicrobial activity of some coumarin esters (1) (Figure 1). These compounds showed good activity against Bacillus sp. and moderate activity against Aspergillus niger. For the data of the antibacterial activity, these compounds were found to be active against Pseudomonas sp. [15].

Figure 1.

Coumarin derivatives with antimicrobial activity.

Al-Amiery et al. have synthesized some coumarin derivatives, and their antifungal activity was determined based on the growth inhibition rates of the mycelia of strains of Aspergillus niger and Candida albicans in Potato Dextrose Broth (PDB) medium against concentrations ranging from 10 to 100 μg/ml. The compound (2) (Figure 1) showed good activity as antifungals against fluconazole as standard drug [16].

Behrami et al. synthesized 8-amino-4,7-dihydroxy-chromen-2-one coumarin derivatives. The antibacterial activities of all the compounds and standard streptomycin and cefalexine at concentrations of 2, 3, and 5 mg/ml were studied against Staphylococcus aureus, Bacillus subtilis, and Escherichia coli. One compound (3) (Figure 1) was more active than cefalexine and lesser active than streptomycin, and it was most active among synthesized compounds [17].

Some coumarin derivatives containing thiazolidin-4-one ring were synthesized by Rama Ganesh et al. and were screened for their antibacterial activity against Gram-positive bacteria Staphylococcus aureus and Bacillus subtilis and Gram-negative bacteria Klebsiella pneumonia and Escherichia coli at the concentration of 0.001 mol/ml compared with the standard drug ciprofloxacin. Zone of inhibition of highly active compound (4) (Figure 1) was 20 mm against Staphylococcus aureus and Bacillus subtilis [18].

1.2 Coumarins as antioxidant agents

Free radicals are molecular species capable of independent existence that contain an unpaired electron in an atomic orbital; they are usually unstable and very reactive. These species are normally produced in the human body from essential metabolic processes, but they may also occur from external sources such as exposure to X-rays, ozone, cigarette smoking, air pollutants, and industrial chemicals [19].

There is an increasing interest in antioxidants, particularly in those intended to prevent the presumed deleterious effects of free radicals in the human body and to prevent the deterioration of fats and other constituents of food stuffs. In both cases, there is a preference for antioxidants from natural rather than from synthetic sources [20]. As improved antioxidant status helps to minimize the oxidative damage and thus delays or prevents pathological changes, potential antioxidant therapy should be included either as natural free-radical-scavenging antioxidant enzymes or as an agent which is capable of augmenting the activity of antioxidant enzymes [21].

The human organism possesses natural systems to annihilate these species, but when the body’s ability to regulate them is overwhelmed, a condition known as oxidative stress appears, free radicals attacking important macromolecules leading to cell damage and homeostatic disruption [22].

Many coumarin derivatives have a special ability to scavenge reactive oxygen species and to influence processes involving free radical injury [23, 24].

Maja et al. synthesized a series of carbohydrazide with coumarin ring (5, 6) and some coumarin derivatives with a heterocyclic ring (7, 8) (Figure 2). All these compounds prove a good antioxidant activity [25].

Figure 2.

Coumarin derivatives with antioxidant activity.

Shivani et al. synthesized new coumarin-substituted derivatives of benzothiazole, and they were evaluated for antioxidant activity by DPPH radical scavenging activity. The test compound (9) (Figure 2) showed good in vitro antioxidant activity [26].

Advertisement

2. Synthesis of coumarin derivatives

Looking to the medicinal importance of the coumarin ring, we employed coumarin as a naturally occurring skeleton for the construction of new derivatives which might exhibit promising antimicrobial and antioxidant activities [27].

The starting materials, 4-methyl/propyl-7-hydroxycoumarin, were prepared by Pechmann synthesis which involved the condensation of resorcinol and ethylacetoacetate/ethylbutyrylacetate in the presence of H2SO4 concentrate [28].

2.1 Mechanism of the Pechmann condensation

The reaction is conducted with a strong Brønsted acid such as methanesulfonic acid or a Lewis acid such as AlCl3. The acid catalyzes transesterification as well as keto-enol tautomerization [29]. A Michael addition leads to the formation of the coumarin skeleton. This addition is followed by rearomatization and then by elimination of water which gives the product (Figure 3).

Figure 3.

Mechanism of the Pechmann condensation.

The coumarin compounds have wide interest due to their diverse pharmacological properties. In particular, these biological activities make coumarin compounds more attractive and testing as novel therapeutic compounds.

As part of our aim in research of biologically active coumarin derivatives, the free hydroxyl group on the coumarin ring has allowed us to introduce some radicals that can improve the biological activity.

The fourth scheme describes the reactions of 4-methyl/propyl-7-hydroxycoumarin with ethyl bromoacetate. 2-Ethyl-((4-methy/propyl-2-oxo-2H-chromen-7-yl)oxy)acetate (IIa–IIb) was prepared by heating a mixture of 4-methyl/propyl-7-hydroxycoumarin and ethyl bromoacetate in the presence of K2CO3 anhydrous in dry acetone. After filtration, the solution was evaporated, and the solid products (IIa or IIb) were recrystallized from ethanol [30] (Figure 4).

Figure 4.

Synthesis of coumarin esters II(a-b).

The fifth scheme describes the reactions of compounds IIa–IIb with hydrazine hydrate. The chemistry of hydrazide and its derivatives has obtained great interest in both organic chemistry and biological science with remarkable impact. Hydrazides and hydrazones are possessing -NH-NH2 and -NH-N=CH- groups, respectively. The availability of proton in hydrazides constitutes them as an important class of compound for new drug discovery. Therefore, researchers have shown great interest in developing these compounds as target structures for evaluating new biological activities [31].

Hydrazinolysis of compounds IIa–IIb gave the corresponding acetohydrazides (IIIa–IIIb) in good yields [30, 32]. One mole of the compound IIa or IIb in ethanol was heated for 4–6 h with hydrazine hydrate (two moles). After this period of time, the mixture was cooled at room temperature, and the precipitate was filtrated and then purified by recrystallization (Figure 5).

Figure 5.

Synthesis of coumarin acetohydrazides III(a-b).

The reaction of the acid hydrazides (4-methyl/propyl-2-oxo-2H-benzopyran-7-oxyacetic acid hydrazide) with CS2 in ethanol containing KOH at room temperature has been presented in Figure 6. The corresponding potassium dithiocarbazate derivatives, IVa–IVb, are obtained [32].

Figure 6.

Synthesis of coumarin potassium salts IV(a-b).

The obtaining of coumarin derivatives with a thiadiazole ring has been described in Figure 7. Literature data show that pyrrole, pyrazole, thiadiazoles, and triazoles and their derivatives are very attractive targets due to their biological properties. In view of the above observations, we have synthesized compounds with thiadiazole ring in order to evaluate the potential antimicrobial and antioxidant activities.

Figure 7.

Synthesis of coumarin thiadiazoles derivatives V(a-b).

These compounds were obtained following the reaction between potassium 4-methyl/propyl-2-oxo-2H-benzopyranyl-7-oxymethyldithiocarbazate and acetic acid. The reaction occurred under refluxing. The solid product was separated by filtration and then purified by recrystallization from acetic acid [33, 34].

In the synthesis of coumarin derivatives, elemental analysis and two basic spectroscopic techniques, infrared spectroscopy (IR) and nuclear magnetic resonance spectroscopy (NMR), were used to characterize the structures of the target compounds [35].

The physical constants and analytical data of compounds Ia–Ib, IIa–IIb, IIIa–IIIb, IVaIVb, and Va–Vb have been given in Tables 15.

Table 1.

Physical constants and analytical data of compounds Ia–Ib.

Table 2.

Physical constants and analytical data of compounds IIa–IIb.

Table 3.

Physical constants and analytical data of compounds IIIa–IIIb.

Table 4.

Physical constants and analytical data of compounds IVa–IVb.

Table 5.

Physical constants and analytical data of compounds Va–Vb.

The IR spectra of all synthesized compounds showed some characteristic peaks indicating the presence of particular groups (Tables 610).

Table 6.

IR spectral data of compounds Ia–Ib.

Table 7.

IR spectral data of compounds IIa–IIb.

Table 8.

IR spectral data of compounds IIIa–IIIb.

Table 9.

IR spectral data of compounds IVa–IVb.

Table 10.

IR spectral data of compounds Va–Vb.

1H-NMR spectra of the synthesized compounds are in accordance with the assigned structures (Table 11). The aliphatic protons resonated in the range of 1.20–4.77 ppm. It can be seen that all the compounds exhibited the respected proton chemical shifts in the same range.

Compd.RChemical shift (δ ppm) 500 MHz
IaH3C▬2.73 ppm s, 3H: CH3; 6.04 ppm s, H3; 6.72 ppm s, H8; 6.83–6.85 ppm d, H6, JH6,H5 = 7.5 Hz; 7.62–7.64 ppm d, H5, JH5,H6 = 7.5 Hz; 10.85 ppm s, 1H: OH
IbH3C▬CH2▬CH21.20 ppm t, 3H: CH3 (12); 1.66 ppm m, 2H: CH2 (11); 2.81–2.84 ppm m, 2H: CH2 (10); 6.11 ppm s, H3; 6.72 ppm s, H8; 6.84–6.85 ppm d, H6, JH6,H5 = 7.5 Hz; 7.63–7.65 ppm d, H5, JH5,H6 = 7.5 Hz; 10.86 ppm s, 1H: OH
IIaH3C▬1.26–1.28 ppm t, 3H: CH3 (14); 2.73 ppm s, 3H: CH3; 4.25–4.28 ppm m, 2H: CH2 (13); 4.61 ppm s, 2H: CH2 (10); 6.04 ppm s, H3; 6.84 ppm s, H8; 7.05–7.07 ppm d H6; 7.62–7.64 ppm d, H5
IIbH3C▬CH2▬CH20.95–0.98 ppm t, 3H: CH3 (17); 1.20–1.23 ppm t, 3H: CH3 (14); 1.60–1.65 ppm m, 2H: CH2 (16); 2.72–2.75 ppm t, 2H: CH2 (15); 4.15–4.20 ppm m, 2H: CH2 (13); 4.92 ppm s, 2H: CH2 (10); 6.17 ppm s, H3; 6.98 ppm s, H8; 6.96–6.97 ppm d H6; 7.73–7.75 ppm d, H5
IIIaH3C▬2.74 ppm s, 3H: CH3; 3.86 ppm s, 2H: NH2 (13); 4.42 ppm s, 2H: CH2 (10); 6.03 ppm s, H3; 6.83 ppm s, H8; 7.05–7.07 ppm d; 7.62–7.64 ppm d, H5; 8.02 ppm s, NH (12)
IIIbH3C▬CH2▬CH20.95–0.98 ppm t, 3H: CH3 (16); 1.59–1.66 ppm m, 2H: CH2 (15); 2.72–2.75 ppm t, 2H: CH2 (14); 4.35 ppm s, 2H: NH2 (13); 4.61 ppm s, 2H: CH2 (10); 6.17 ppm s, H3; 6.98 ppm s, H8; 6.99–7.00 ppm d H6; 7.74–7.76 ppm d, H5; 9.42 ppm s, NH (12)
IVaH3C▬2.71–2.74 ppm d, 3H: CH3; 4.41–4.43 ppm s, 2H: CH2 (10); 6.02–6.05 ppm q, H3; 6.82–6.84 ppm d, H8; 7.05–7.08 ppm q H6; 7.62–7.64 ppm d, H5; 9.94 ppm s, NH (12); 11.23 ppm s, NH (13)
IVbH3C▬CH2▬CH21.20–1.22 ppm t, 3H: CH3 (18); 1.62–1.66 ppm m, 2H: CH2 (17); 2.79–2.82 ppm t, 2H: CH2 (16); 4.40 ppm s, 2H: CH2 (10); 6.10 ppm s, H3; 6.84 ppm s, H8; 7.04–7.08 ppm q H6; 7.62–7.66 ppm d, H5; 9.95 ppm s, NH (12); 11.25 ppm s, NH (13)
VaH3C▬2.39 ppm s, 3H: CH3; 4.77 ppm s, 2H: CH2 (10); 6.23 ppm s, H3; 6.99 ppm s, H8; 7.02–7.04 ppm d H6; 7.70–7.72 ppm d, H5; 10.29 ppm s, SH (16)
VbH3C▬CH2▬CH20.95–0.98 ppm t, 3H: CH3 (19); 1.60–1.65 ppm m, 2H: CH2 (18); 2.72–2.75 ppm t, 2H: CH2 (17); 4.77 ppm s, 2H: CH2 (10); 6.17 ppm s, H3; 6.98 ppm s, H8; 6.99–7.00 ppm d H6; 7.75–7.76 ppm d, H5; 11.27 ppm s, SH (16)

Table 11.

1H-NMR spectral data of compounds I–V.

Advertisement

3. Pharmacological activities of coumarin derivatives

3.1 Antimicrobial activity

The compounds were screened for their antibacterial and antifungal activity according to standard protocols [36].

The antimicrobial activity was studied using Gram-positive bacteria (Staphylococcus aureus ATCC 25923, Sarcina lutea ATCC 9341, Bacillus cereus ATCC 14579), Gram-negative bacteria (Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853), and pathogenic yeasts (Candida albicans ATCC 10231, Candida glabrata ATCC MYA 2950, Candida parapsilosis ATCC 22019). All these strains were obtained from the Culture Collection of the Department of Microbiology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania.

Antimicrobial activity was evaluated by agar disc diffusion method (CLSI, 2014). A small amount of each microbial culture was diluted in sterile 0.9% NaCl until the turbidity was equivalent to McFarland standard no. 0.5 (106 CFU/ml). The suspensions were further diluted 1:10 in Mueller-Hinton agar for bacteria and Sabouraud agar for yeasts and then spread on sterile Petri plates (25 ml/Petri plate). Sterile stainless steel cylinders (5 mm internal diameter; 10 mm height) were applied on the agar surface in Petri plates. Then, 0.1 ml of each compound (10 mg/ml in DMSO) was added into the cylinders. The DMSO solvent was also tested in order to assess its intrinsic antimicrobial activity. Commercial available discs containing ampicillin (25 μg/disc), chloramphenicol (30 μg/disc), and nystatin (100 μg/disc) were also placed on the agar surface. The plates were incubated at 37°C for 24 h (bacteria) and at 24°C for 48 h (yeasts). After incubation the diameters of inhibition zones were read in triplicate. Statistical analysis of the results included the calculation of standard deviation (Tables 12 and 13).

Compd./referenceDiameters of the growth inhibition zone (mm)
S. aureus ATCC 25923S. lutea ATCC 9341B. cereus ATCC 14579E. coli ATCC 25922Pseudomonas aeruginosa
ATCC 27853
Ia14 ± 0.5225 ± 0.7925 ± 1.5212 ± 0.798 ± 0.93
Ib27 ± 1.2929 ± 0.83NANANA
IIa14 ± 0.9122 ± 0.7926 ± 0.7911 ± 0.528 ± 1.43
IIb27 ± 0.5230±NANANA
IIIa15 ± 0.5420 ± 0.7922 ± 0.7910 ± 0.799 ± 0.79
IIIb25 ± 0.5228±NANANA
IVa17 ± 1.0825 ± 0.9124 ± 1.5212 ± 0.939 ± 1.43
IVb25 ± 1.0830 ± 0.83NANANA
Va14 ± 0.5225 ± 0.5220 ± 1.4310 ± 1.528 ± 0.52
Vb21 ± 1.4325 ± 0.79NA13 ± 0.83NA
Ampicillin
(25 μg/disc)
26 ± 0.0436 ± 0.00NA21 ± 0.79NA
Chloramphenicol (30 μg/disc)22 ± 0.0038 ± 0.0024 ± 0.0021 ± 0.52NA

Table 12.

Antibacterial activity of compounds I–V.

Data are mean ± SD (n = 3); NA, no activity.

Compd./referenceDiameters of the growth inhibition zone (mm)
C. albicans
ATCC 10231
C. glabrata
ATCC MYA 2950
C. parapsilosis
ATCC 22019
Ia24 ± 1.8321 ± 0.5234 ± 1.83
Ib10 ± 0.9110 ± 0.7910 ± 0.54
IIa25 ± 0.5227 ± 0.5435 ± 1.83
IIb9 ± 1.83NANA
IIIa19 ± 1.7924 ± 0.5224 ± 1.79
IIIb12 ± 1.8311 ± 0.5411 ± 0.54
IVa23 ± 0.9116 ± 0.5225 ± 1.08
IVb10 ± 0.5412 ± 1.08NA
Va16 ± 1.7921 ± 1.8321 ± 0.54
Vb9 ± 0.549 ± 0.52NA
Nystatin (100 μg/disc)25 ± 0.5225 ± 0.5224 ± 0.00

Table 13.

Antifungal activity of compounds I–V.

Data are mean ± SD (n = 3); NA, no activity.

The qualitative screening of the antimicrobial activity was performed in order to identify the antimicrobial spectrum of the tested compounds. The inhibitory effects of the synthetic compounds against Gram-positive and Gram-negative bacteria and fungi are given in Tables 12 and 13 [35].

According to the results of the antibacterial studies, the efficacy of the tested compounds against Gram-positive bacteria was higher than that exhibited for Gram-negative bacteria. All the synthesized compounds were very active against S. aureus ATCC 25923, the most active compounds being Ib, IIb, IIIb, and IVb. The replacement of the methyl radical in the fourth position with the propyl group was correlated with an increased activity against S. aureus ATCC 25923.

The tested compounds exhibited excellent antibacterial activity against S. lutea, the most active derivatives being IIb, IVb, Ib, and IIIb.

We found a moderate action against B. cereus ATCC 14579, the most active being the umbelliferone derivatives with a methyl group attached to C4: IIa, Ia, and IVa.

Against Escherichia coli ATCC 25922, the investigated compounds had a weaker action than the controls ampicillin and chloramphenicol. The most active was the compound that contains a thiadiazole ring, Vb.

The presence of the methyl group attached to the coumarin ring in the fourth position had a positive influence on the anti-Pseudomonas ATCC 27853 potential of the compounds, all the tested 4-propyl-coumarin derivatives being inactive.

We have noticed a very important action against the investigated Candida strains; all tested compounds were found to be very active against fungi. The compounds IIa and Ia had a greater inhibitory potential against C. parapsilosis ATCC 22019 than nystatin. The introduction of the sulfur atom appeared to be correlated with a good anti-Candida activity.

3.2 Antioxidant activity

In order to evaluate the antioxidant activity of synthesized compounds, we use three antioxidant assays: DPPH radical inhibition, total reducing power, and nitric oxide (NO) inhibition.

The DPPH assay is based on assessing the substances’ ability to reduce the stable radical (diphenylpicrylhydrazyl) to diphenylpicrylhydrazine. The DPPH free radical, bearing an odd electron, gives a strong absorption maximum at λ = 517 nm (purple color). When the odd electron of the DPPH radical pairs with a hydrogen atom from an antioxidant, the reduced form DPPH-H is created, and the color turns from purple to yellow [36, 37].

A possible mechanism that can explain the antioxidant effect of the coumarin hydrazide derivatives is related to the keto-enol forms of the substances, the enol group being capable to easily donate the hydrogen (Figure 8) [38].

Figure 8.

Proposed mechanism for antioxidant activity of coumarin hydrazides.

The experimental procedure for the DPPH assay was adapted from literature [27, 28, 37], only slight modifications being made. Briefly, 2.5 ml solution of DPPH (2,2-diphenyl-1-picrylhydrazyl) radical 0.1 mM in methanol was added over 0.5 ml of methanolic solution of the tested compound (1 mg/ml). The absorbance of the DPPH solution at 517 nm was determined spectrophotometrically before (Acontrol) and 15 minutes after adding the solutions of the compounds (Atest), and the percentage of activity was calculated. Ascorbic acid was used as a reference compound:

%radical scavenging activity=AcontrolAtest×100/Acontrol

where Acontrol is the absorbance of the control sample (DPPH solution without test sample) and Atest is the absorbance of the test sample (DPPH solution + test compound).

Out of the tested compounds, the most active DPPH free radical scavengers were the coumarin hydrazide derivatives (IIIa–IIIb, IVa–IVb). The activities of IVa were similar to that of the standard, the inhibition percentage being over 90%, the introduction of sulfur atoms in the molecule having a positive influence on the scavenging potential. Compounds IIIa and IVa, containing a methyl group, were slightly more active than their analogues with propyl radical (Table 14) [19].

Compd.Inhibition percentage (%)
Ia21.4
Ib21.2
IIa19
IIb19.83
IIIa73
IIIb53.56
IVa95
IVb87.67
Va31.51
Vb25.13
Ascorbic acid
1 mg/ml
96.8

Table 14.

DPPH inhibition percentages of compounds I–V (1 mg/ml).

Fe(III) reduction is often used as an indicator of electron-donating activity. In the reducing power assay, antioxidants with electron-donating abilities reduce ferricyanide to ferrocyanide by donating an electron. The amount of ferrocyanide is monitored by measuring the formation of Perl’s Prussian blue at 700 nm. Increasing absorbance at 700 nm indicates an increase in reducing ability [39]. Within this assay, EC50 values are the effective concentrations at which the absorbance is 0.5.

The solution of the test compound (0.5 ml) at different concentrations in methanol was mixed with phosphate buffer (1.25 ml, 0.2 mol/l, pH 6.6) and potassium ferricyanide 1% (1.25 ml), and the mixture was incubated at 50°C for 20 min. At the end of the incubation period, trichloroacetic acid 10% (1.25 ml) was added to the mixture and centrifuged at 3000 rpm for 10 min. The upper layer solution was collected, and 2.5 ml were mixed with distilled water (2.5 ml) and ferric chloride 0.1% (0.5 ml). The absorbance was measured after 15 min at 700 nm against a blank. The EC50 values were calculated by linear interpolation between values above and below 50% activity. Ascorbic acid was used as reference [28, 30, 36, 37].

The reducing power of the tested compounds was modest, and the results are presented in Table 15. The only substances that were moderately active were the hydrazide derivatives IIIa and IVa, but their activity was inferior to that exhibited by the reference substance (ascorbic acid) [19].

Compd.Extinction
Ia0.0599
Ib0.0105
IIa0.0234
IIb0.0010
IIIa(1 mg/ml)1.3432
(0.8 mg/ml)1.1237
(0.6 mg/ml)1.0821
(0.4 mg/ml)0.9674
(0.2 mg/ml)0.5499
IVa(1 mg/ml)0.6592
(0.8 mg/ml)0.5900
(0.6 mg/ml)0.4854
(0.4 mg/ml)0.4258
(0.2 mg/ml)0.2785
IVb0.4887
Va0.0472
Vb0.3063
Ascorbic acid
1 mg/ml
2.8261

Table 15.

The reducing power of compounds I–V.

The calculated values for EC50 are shown in Table 16. This method could not be applied to compound IIIb due to the formation of an abundant precipitate in the process.

Compd.EC50 (mg/ml)
Ia>>1
Ib>>1
IIa>>1
IIb>>1
IIIa0.176
IVa0.627
IVb1.02
Va>>1
Vb>1
Ascorbic acid
1 mg/ml
0.049

Table 16.

The calculated values of EC50.

Nitric oxide is involved in a variety of biological functions (neurotransmission, vascular homeostasis, antimicrobial and antitumor activities). NO was primarily described as a regulator of vascular tones in the cardiovascular system. Beyond this function it can prevent platelet activation, limit leukocyte adhesion to the endothelium, and regulate myocardial contractility, and it is involved in immune system reactions.

Despite the possible beneficial effects of NO, it also contributes to oxidative damage. In general, the overwhelming production of NO contributes to the pathogenesis of both acute and chronic inflammatory processes, and NO has been recognized as one of the main signaling molecules involved in these processes [23, 40]. Therefore, compounds that act like nitric oxide inhibitors have beneficial effects.

The NO inhibition assay is based on the diazotization of sulfanilic acid at acid pH by nitric oxide. The reaction product is subsequently coupled stoichiometrically with N-(1-naphthyl)ethylenediamine, forming a colored azo compound which is measured spectrophotometrically at a peak absorbance of 548 nm [36].

0.5 ml of the tested coumarin derivative solution, as well as ascorbic acid (standard compound), was taken in separate tubes, and 2.0 ml of sodium nitroprusside (10 mM) and 0.5 ml phosphate buffer saline (pH = 7.4) were added to each tube. The solutions were incubated at 25°C for 150 minutes. After the incubation, over 0.5 ml of the incubated solution 1 ml of sulfanilic acid 0.33% was added, and the mixture was left for 5 min at room temperature; after this period of time, 1 ml naphthylethylene diamine (NED) HCl reagent 0.1% was added, and the solutions were incubated for another 30 min. The absorbance was measured at 546 nm [38].

Most of the investigated compounds were moderate NO inhibitors (Table 17) [19].

Compd.NO inhibition (%)
Ia15.2
Ib19.8
IIa12.11
IIb14.7
IIIa22.8
IIIb55.5
IVa28.45
IVb29.44
Va22.6
Vb26
Ascorbic acid
50 mg/ml
77.19

Table 17.

NO inhibition activity of compounds I–V.

Advertisement

4. Concluding remarks

We have synthesized some coumarin derivatives starting from 4-methyl-7-hydroxycoumarin with antimicrobial and antioxidant activities to different reaction steps. The IR and NMR spectra of the synthesized compounds were in accordance with the assigned structures. All the synthesized compounds were very active against S. aureus ATCC 25923, and they exhibited excellent antibacterial activity against S. lutea. The presence of the methyl group attached to the coumarin ring in the fourth position had a positive influence on the anti-Pseudomonas ATCC 27853 potential of the compounds, all the tested 4-propyl-coumarin derivatives being inactive. Against the investigated Candida strains, all tested compounds were found to be very active. The introduction of the sulfur atom appeared to be correlated with a good anti-Candida activity. The most active DPPH free radical scavengers were the coumarin hydrazide derivatives, the activities of these being similar to that of the standard. The reducing power of the tested compounds was modest, and only the hydrazide derivatives were moderately active. Most of the investigated compounds were moderate NO inhibitors.

The interest in the synthesis of coumarin derivatives has been gaining importance over the last decades, reflecting the importance of such compounds in both medical and chemical research. Future goals for this field of research include the discovery, synthesis, and development of compounds which display increased potency, as well as fueling structure–activity relationship studies aimed at understanding the modes of action of the most biologically active members of these classes of products.

Although coumarin is a simple molecule and many of its derivatives have been known for more than a century, it continues to maintain the interest of researchers being a plentiful source of potential drug candidate because of their significant therapeutic potential.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Raev L, Voinov E, Ivanov I, Popov D. Antitumor activity of some coumarin derivatives. Pharmazie. 1990;45(9):696-698
  2. 2. Nofal ZM, El-Zahar M, Abd El-Karim S. Novel coumarin derivatives with expected biological activity. Molecules. 2000;5(2):99-113. DOI: 10.3390/50200099
  3. 3. El-Agrody AM, Abd El-Latif MS, El-Hady NA, Fakery AH, Badair AH. Heteroaromatisation with 4-hydroxycoumarin part 2. Molecules. 2001;6(6):519-527. DOI: 10.3390/60600519
  4. 4. Patibha S, Shreeya P. Synthesis, characterization and antimicrobial studies of some novel 3-arylazo-7-hydroxy-4-methylcoumarin. Indian Journal of Chemistry. 1999;38B:1139-1142
  5. 5. Patonay T, Litkei GY, Bognar R, Erdei J, Misztic C. Synthesis, antimicrobial and antifungal activity of 4-hydroxycoumarin derivatives, analogues of Novobiocin. Pharmazie. 1984;39:86-91
  6. 6. Shaker RM. Synthesis and reactions of some new 4H-pyrano[3,2-c]benzopyran-5-one derivatives and their potential biological activities. Pharmazie. 1996;51(3):148-151
  7. 7. Cacic M, Molnar M, Balic T, Draca N, Rajkovic V. Design and Synthesis of Some Thiazolidin-4-ones Based on (7-Hydroxy-2-oxo-2H-chromen-4-yl) Acetic Acid. Molecules. 2009;14:2501-2513. DOI: 10.3390/molecules14072501
  8. 8. Manolov I, Maichle-Moessmer C, Danchev N. Synthesis, structure, toxicological and pharmacological investigations of 4-hydroxycoumarin derivatives. European Journal of Medicinal Chemistry. 2006;41(7):882-890. DOI: 10.1016/j.ejmech.2006.03.007
  9. 9. Al-Majedy Y, Al-Amiery A, Kadhum AA, Bakar Mohamad A. Antioxidant activity of coumarins. Systematic Reviews in Pharmacy. 2017;8(1):24-30. DOI: 10.5530/srp.2017.1.6
  10. 10. Emmanuel-Giota AA, Fylaktakidou KC, Hadjipavlou-Latina DJ, Litinas KE, Nicolaides DN. Synthesis and biological evaluation of several 3-(coumarin-4-yl)tetrahydroisoxazole and 3-(coumarin-4-yl)dihydropyrazole derivatives. Journal of Heterocyclic Chemistry. 2001;38(3):717-722. DOI: 10.1002/jhet.5570380329
  11. 11. Tenover FC. Mechanisms of antimicrobial resistance in bacteria. The American Journal of Medicine. 2006;119(6):S3-S10. DOI: 10.1016/j.amjmed.2006.03.011
  12. 12. Rookaya M, Karenchak L, Romanowski E, Kowalski R. Fourth generation fluoroquinolones: New weapons in the arsenal of ophthalmic antibiotics. American Journal of Ophthalmology. 2002;133(4):463-466
  13. 13. Veselinović J, Matejić J, Veselinović A, Sokolović D. Coumarin structure as a lead scaffold for antibacterial agents - molecular docking study. Biologica Nyssana. 2016;7(2):167-170. DOI: 10.5281/zenodo.200417
  14. 14. Al-Majedy Y, Al-Amiery A, Kadhum AA, Bakar Mohamad A. Coumarins: The antimicrobial agents. Systematic Reviews in Pharmacy. 2017;8(1):62-70. DOI: 10.5530/srp.2017.1.11
  15. 15. Medimagh-Saidana S, Romdhane A, Daami-Remadi M, Jabnoun-Khiareddine H, Touboul D, Ben Jannet H, et al. Synthesis and antimicrobial activity of novel coumarin derivatives from 4-methylumbelliferone. Medicinal Chemistry Research. 2015;24:3247-3257. DOI: 10.1007/s00044-015-1368-y
  16. 16. Al-Amiery AA, Kadhum AAH, Mohamad AB. Antifungal activities of new coumarins. Molecules. 2012;17:5713-5723. DOI: 10.3390/molecules17055713
  17. 17. Behrami A, Krasniqi I. Antibacterial activity of coumarine derivatives synthesized from 8- amino-4,7-dihydroxy-chromen-2-one and comparison with standard drug. Journal of Chemical and Pharmaceutical Research. 2012;4(5):2495-2500
  18. 18. RamaGanesh CK, Yadav DB, Venkatesh KB. Synthesis and biological evaluation of some innovative coumarin derivatives containing thiazolidin-4-one ring. Indian Journal of Chemistry. 2010;49B:1151-1154
  19. 19. Tataringa G, Stan C, Mircea C, Jitareanu A, Zbancioc AM. Antioxidant evaluation of some coumarin derivatives. Farmácia. 2016;64(4):533-538
  20. 20. Yasameen K, Al-Majedy D, Al-Duhaidahawi K, Al-Azawi A, Kadhum A, Mohamad AB. Coumarins as potential antioxidant agents complemented with suggested mechanisms and approved by molecular modeling studies. Molecules. 2016;21:135-146. DOI: 10.3390/molecules21020135
  21. 21. Bast A, Haenen GR, Doelman CJ. Oxidants and antioxidants: State of the art. The American Journal of Medicine. 1991;91(3):S3-S13. DOI: 10.1016/0002-9343(91)90278-6
  22. 22. Lobo V, Patil A, Phatak A, Chandra N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacognosy Reviews. 2010;4(8):118-126. DOI: 10.4103/0973-7847.70902
  23. 23. Patel RM, Patel NJ. In vitro antioxidant activity of coumarin compounds by DPPH, super oxide and nitric oxide free radical scavenging methods. Journal of Advanced Pharmacy Education & Research. 2011;1:52-68
  24. 24. Jamier V, Marut W, Valente S, Chereau C, Chouzenoux S, Nicco C, et al. Chalcone-Coumarin derivatives as potential anti-cancer drugs: An in vitro and in vivo investigation. Anti-Cancer Agents in Medicinal Chemistry. 2014;14(7):963-974. DOI: 10.2174/1871520613666131224124445
  25. 25. Maja M, Čačić C. Antioxidant activity of some (7-hydroxy-2-oxo-2H-chromen-4yl) acetic acid derivatives. Croatian Journal of Food Science and Technology. 2012;4(1):54-63
  26. 26. Gulati S, Wakode S, Kaur A, Anand K. Synthesis, biological activity and recent advancement of benzothiazoles: A Classical Review. World Journal of Pharmacy and Pharmaceutical Sciences. 2017;6(6):1842-1869. DOI: 10.20959/wjpps20176-9423
  27. 27. Mohamed HM, Abd EL-Wahab HF, EL-Agrody AM, Bedair AH, Eid FA, Khafagy MM, et al. Synthesis and characterization of new diiodocoumarin derivatives with promising antimicrobial activities. Beilstein Journal of Organic Chemistry. 2011;7:1688-1696. DOI: 10.3762/bjoc.7.199
  28. 28. Hamdi N, Al-Ayed AS, Said RB, Fabienne A. Synthesis and characterization of new thiazolidinones containing coumarin moieties and their antibacterial and antioxidant activities. Molecules. 2012;17(8):9321-9334. DOI: 10.3390/molecules17089321
  29. 29. Sreeja S, Arul K, Kumaran J. New Coumarin Derivatives. Design, Synthesis and Pharmacological Evaluation. LAP LAMBERT Saarbrücken, Deutschland/Germany: Academic Publishing; 2013. 5p
  30. 30. Satyanarayana VSV, Sreevani P, Sivakumar A, Vijayakumar V. Synthesis and antimicrobial activity of new Schiff bases containing coumarin moiety and their spectral characterization. Arkivok. 2008;xvii:221-223. DOI: 10.3998/ark.5550190.0009.h21
  31. 31. Khan MS, Siddiqui SP, Tarannum N. A systematic review on the synthesis and biological activity of hydrazide derivatives. Hygeia Journal for Drugs and Medicine. 2017;9(1):61-79. DOI: 10.15254/H.J.D.Med.9.2017.165
  32. 32. Upmanyu N, Kumar S, Shah K, Mishra P. Synthesis and antimicrobial studies of some 4-(substituted)-ethanoylamino 3-mercapto-5-(4- substituted) phenyl-1,2,4-triazoles. Dhaka University Journal of Pharmaceutical Sciences. 2012;11(1):7-18. DOI: 10.3329/dujps.v11i1.12481
  33. 33. El-Hashash MA, Guirguis DB, Abd El-Wahed NAM, Kadhim MA. Synthesis of novel series of phthalazine derivatives with antibacterial and antifungal evaluation. Journal of Chemical Engineering & Process Technology. 2014;5(4):1-8. DOI: 10.4172/2157-7048.1000195
  34. 34. Rehman A, Siddiqui SZ, Abbasi MA, Abbas N, Khan KM, Shahid M, et al. Synthesis, antibacterial screening and hemolytic activity of s-substituted derivatives of 5-benzyl-1,3,4-oxadiazole-2-thiol. International Journal of Pharmacy and Pharmaceutical Sciences. 2012;4(2):676-680
  35. 35. Tataringa G, Tuchilus C, Jitareanu A, Stan CD, Zbancioc AM. Antimicrobial prospection of some coumarin derivatives. Farmácia. 2016;66(2):323-330
  36. 36. Ranković B, Kosanić M, Stanojković T. Stereocaulon paschale lichen as antioxidant, antimicrobial and anticancer agent. Farmácia. 2014;62(2):306-317
  37. 37. Kadhum AAH, Ahmed AA, Musa AY, Mohamad AB. The antioxidant activity of new coumarin derivatives. International Journal of Molecular Sciences. 2011;12(9):5747-5761. DOI: 10.3390/ijms12095747
  38. 38. Matthews EK, Seaton ED, Forsyth MJ, Humphrey PPA. Photon pharmacology of an iron-sulphur cluster nitrosyl compound acting on smooth muscle. British Journal of Pharmacology. 1994;113(1):87-94
  39. 39. Kenchappa R, Bodke YD, Chandrashekar A, Sindhe MA, Peethambaret SK. Synthesis of coumarin derivatives containing pyrazole and indenone rings as potent antioxidant and antihyperglycemic agents. Arabian Journal of Chemistry. 2017;10(2):S3895-S3906. DOI: 10.1016/j.arabjc.2014.05.029
  40. 40. Víteček J, Lojek A, Valacchi G, Kubala L. Arginine-based inhibitors of nitric oxide synthase: Therapeutic potential and challenges. Mediators of Inflammation. 2012;2012:1-22. DOI: 10.1155/2012/318087

Written By

Gabriela Tataringa and Ana Maria Zbancioc

Submitted: 04 May 2019 Reviewed: 17 June 2019 Published: 27 July 2019