Int J Pharm Pharm Sci, Vol 8, Issue 1, 52-61Original Article


MOLECULAR DOCKING STUDY OF FLAVONOID COMPOUNDS AS INHIBITORS OF Β-KETOACYL ACYL CARRIER PROTEINSYNTHASE II (KAS II) OF PSEUDOMONAS AERUGINOSA

GHALIA SABBAGH*, NOURA BERAKDAR**

Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Aleppo Aleppo University Street, Aleppo, Syria
Email: ghaliaaa@hotmail.com

 Received: 30 Jun 2015 Revised and Accepted: 18 Nov 2015


ABSTRACT

Objective: Fatty acid biosynthesis is essential for bacterial survival. Components of this biosynthetic pathway have been identified as attractive targets for the development of new antibacterial agents. β–Ketoacyl acyl carrier protein synthase (KAS) II is a key catalyst in bacterial fatty acid biosynthesis. It is related to control the temperature dependent regulation of fatty acid composition.

Methods: Structure of KasII (FabF) was retrieved from the Protein Data Bank and the structures of flavonoid compounds have been collected from zinc database. Molecular docking and drug likeness studies were performed for those natural compounds to evaluate and analyze the anti-antimicrobial activity.

Results: Finally one compound, Casticin binds to KAS II with the most favorable binding energy (-112.5 kcal/mol) whereas the reference (-92.76 kcal/mol). The fitness score of the compound suggest that this lead can be formulate as an antimicrobial activities drug against gram-negative Pseudomonas aeruginosa.

Conclusions: The results of this study can be implemented in vitro and in vivo in the drug designing pipeline.

Keywords:Pseudomonas aeruginosa, Fatty acid synthases, KAS II, Docking, Flavonoids, iGEMDOCK.


INTRODUCTION

Pseudomonas aeruginosa is a Gram-negative, aerobic; a non-fermentative bacterium widely distributed in nature and can survive on a wide variety of surfaces and among hospital environment [1]. Pseudomonas aeruginosa is a common nosocomial pathogen that causes a variety of infections and is an opportunistic human pathogen. It is “opportunistic” because it seldom infects healthy individuals. Instead, it often colonizes immunocompromised patients, like those with cystic fibrosis, cancer or AIDS [2, 3]. The most common cause of burn infections is P. aeruginosa. It is such a potent pathogen that it attacks up two thirds of the critically ill-hospitalized patients and this usually portends more invasive diseases and acute diseases caused by P. aeruginosa tend to be chronic and life-threatening [4]. P. aeruginosa develops resistance by various mechanisms like multi-drug resistance efflux pumps, biofilm.

Formation, production of β-lactamases and aminoglycoside modifying enzymes [5,6]. Although several classes of antibacterial agents are presently available, resistance in most of the pathogenic bacteria to these drugs constantly emerges. In order to prevent this serious medical problem, the elaboration of new types of antibacterial agents or the expansion of bioactivity of the previous drugs is a very important task [7]. Therefore, in recent years, the research has been focused toward development of new antibacterial agents, which may act through novel target, overcoming the problem of acquired resistance.

A promising target is the fatty acid synthase (FAS) pathway in bacteria. Fatty acid biosynthesis (FAB) is an essential metabolic process of prokaryotic organisms and is required for cell viability and growth [8]. Large multifunctional proteins termed type I fatty acid synthases (FAS I) catalyze these essential reactions to eukaryotes [9, 10]. In contrast, bacteria use multiple enzymes to accomplish the same goal and are referred to as type II, or dissociated, fatty acid synthases (FAS II) [11]. The type II system has been most extensively studied in Pseudomonas aeruginosa [12, 13]. The β-ketoacyl carrier protein synthases (β-KAS) are key regulators of fatty acid biosynthesis. It is well known that three types of β-ketoacyl acyl carrier protein synthase (KAS) enzymes, KAS I (FabB, β-ketoacyl–ACP synthase I), KAS II (FabF, β-ketoacyl–ACP synthase II), and KAS III (FabH, β-ketoacyl–ACP synthase III).

The β-Ketoacyl-Acyl Carrier Protein Synthase I (KAS I) play a very important role in the elongation of fatty acids. Studies have shown that mutants lacking the KAS I enzymes face serious problems in growth and require exogenous unsaturated fatty acids. The function of KAS II is to mainly control the thermal regulation of fatty acid composition. Lack of KAS II will result in failure of the elongation of palmitoleate to cis vaccinate. However, under standard culture conditions, growth is not suppressed. Finally, KAS III is responsible for controlling the rate of fatty acid syntheses, by catalyzing the first step in the pathway, fig. 1.

Fig. 1: The bacterial type II fatty acid biosynthetic pathway [72]

These enzymes catalyze the Claisen condensation reaction, transferring an acyl primer to malonyl-ACP (Acyl Carrier Protein) and thereby creating a β-ketoacyl–ACP that has been lengthened by two carbon units [14].

Flavonoids are becoming the subject of medical research and they have been reported to possess many useful properties, including anti-inflammatory, oestrogenic, enzyme inhibition, antimicrobial [15, 16], antiallergic, antioxidant [17], vascular and cytotoxic antitumour activities. The basic structural feature of flavonoid compounds is the 2-phenyl-benzo pyrane or flavane nucleus, which consists of two benzene rings (A and B) linked through a heterocyclic pyrane ring (C) [18], fig. 2.

Fig. 2: The skeleton structure of the flavones (a class of flavonoids), with rings named and positions numbered

They can be further classified by their chemical structures that are flavones, flavonols, flavanols, flavanones, isoflavones, and anthocyanins. The in silico method is used to analyze the target of structures of expecting to bind sites, to generate candidate molecules, to check for their drug likeness, to dock them with the target, to rank them according to their binding energies, and further to optimize the molecules for improving binding the features.

In this work, we computationally predict that flavonoid casticincan be used as potential drug candidates against Gram-negative Pseudomonas aeruginosa based on that good binding energy toward KAS II active sites.

MATERIALS AND METHODS

Protein preparation

The protein, required for the docking studies, has been retrieved from the Protein Data Bank (PDB) [19]. The protein has (4JPF) a resolution factor of 1.67 Å [19].

We defined the active site of KAS II based on the x-ray complex structure of KAS II protein and 3-benzamido-2-hydroxybenzoic acid ligand. The binding sites, which are more flexible, were selected for this study.

Chemical structures were retrieved from ZINC database [20]. The MOL2 structural formats of all the 50 components were generated from the database ZINC. The set of ligand molecules selected for this study was 50 flavonoids compounds from different plant sources and which have been selected after an extensive literature survey that was performed to hunt for flavonoids that have antimicrobial activities via pubmed site [21-26].

The literature on flavonoids and antimicrobial activities has been collected from this database. The flavonoids have aroused considerable interest recently because of their potentially beneficial effects on human health. They have been reported to have anti-microbial, anti-allergic, anti-platelet, anti-inflammatory, anti-tumor and anti-oxidant activities [27-61] and many flavonoids display low toxicity [62] in mammals. In fact, we have selected 50 flavonoids without sugars (only the aglycones), as the anti-bacteria effectiveness increased when the sugar is separated [62]. Table1 is shown the chemical structure of 50 flavonoids with the reference. They were also selected in accordance with the Lipinski’s rules of five.

Table 1: The structure of 50 screened flavonoid compounds used in this study

S. No.

Compound ID

Chemical name

IUPAC Name

Compound structure

References

1

Zinc_6018556

Casticin

5-hydroxy-2-(3-hydroxy-4-methoxyphenyl)-

3,6,7-trimethoxy-4H-chromen-4-one

35

2

zinc_27646615

Tangeritin

5,6,7,8-tetramethoxy-2-(4-methoxyphenyl)

chromen-4-one

60

3

Zinc_6484604

Tamarixetin

3,5,7-Trihydroxy-2-(3-hydroxy-4-methoxyphenyl)-4-benzopyrone

33

4

Zinc_897714

Malvidin

3,5,7-trihydroxy-2-(4-hydroxy-3,5-dimethoxyphenyl)chromenium

28

5

Zinc_517261

Isorhamnetin

3,5,7-trihydroxy-2-(4-hydroxy-3-

methoxyphenyl)chromen-4-one

28

6

Zinc_6483609

Syringetin

3,5,7-trihydroxy-2-(4-hydroxy-3,5-dimethoxy-phenyl)-chromen-4-one

45

7

Zinc_3954302

Petunidin

2-(3,4-dihydroxy-5-methoxyphenyl)-

3,5,7-trihydroxychromenium

28

8

Zinc_5998961

Tricin

5,7-dihydroxy-2-(4-hydroxy-3,5-

dimethoxyphenyl)chromen-4-one

39

9

Zinc-39111

Fisetin

2-(3,4-dihydroxyphenyl)-3,7-

dihydroxychromen-4-one

27. 28

10

Zinc_6536276

Herbacetin

3,5,7,8-tetrahydroxy-2-(4-hydroxyphenyl)

chromen-4-one

34

11

Zinc_14728065

Cajanin

3-(2,4-dihydroxyphenyl)-5-hydroxy-7-

methoxychromen-4-one

46

12

Zinc_5733553

Pectolinarigenin

5,7-dihydroxy-6-methoxy-2-(4-methoxyphenyl)

chromen-4-one

48

13

Zinc_4098322

Homoeriodictyol

5,7-dihydroxy-2-(4-hydroxy-3-methoxyphenyl)-2,3-dihydrochromen-4-one

32

14

Zinc_1081534

Sternbin

2-(3,4-dihydroxyphenyl)-5-hydroxy-

7-methoxy-2,3-dihydrochromen-4-one

36

15

Zinc_18185774

Luteolin

2-(3,4-dihydroxyphenyl)-5,7-

dihydroxychromen-4-one

28

16

Zinc_5732241

Hispidulin

5,7-dihydroxy-2-(4-hydroxyphenyl)-6-

methoxychromen-4-one

37

17

zinc_1429478

Benzoic acid,

3-(benzoylamino)-2-hydroxy

3-benzamido-2-hydroxybenzoic acid

61

18

Zinc_14807049

Onysilin

5-hydroxy-6,7-dimethoxy-2-phenyl-

2,3-dihydrochromen-4-one

57

19

Zinc_39091

Hesperetin

5,7-dihydroxy-2-(3-hydroxy-4-methoxyphenyl)-

2,3-dihydrochromen-4-one

28

20

Zinc_161951

Naringenin trimethyl ether

5,7-dimethoxy-2-(4-methoxyphenyl)-

2,3-dihydrochromen-4-one

47

21

Zinc_4098238

butin

2-(3,4-dihydroxyphenyl)-7-hydroxy-

2,3-dihydro-4H-chromen-4-one

28

22

Zinc_14728050

Steppogenin

2-(2,4-dihydroxyphenyl)-5,7-dihydroxy-

2,3-dihydrochromen-4-one

31

23

Zinc_57857

6,2',4'-Trimethoxyflavanone

2-(2,4-dimethoxyphenyl)-6-methoxy-

2,3-dihydro-4H-chromen-4-one

54

24

Zinc_5998641

dihydrooroxylin A

5,7-dihydroxy-6-methoxy-2-phenyl-

2,3-dihydrochromen-4-one

38

25

Zinc_3869768

Kaempferol

3,5,7-trihydroxy-2-(4-hydroxyphenyl)

chromen-4-one

27. 28

26

Zinc_5733652

Diosmetin

5,7-dihydroxy-2-(3-hydroxy-4 methoxyphenyl)

chromen-4-one

42

27

Zinc_2146973

Isosakuranetin

5,7-dihydroxy-2-(4-methoxyphenyl)-2,3-

dihydrochromen-4-one

43

28

Zinc_1561069

Naringenin 7,4'-dimethyl ether

5-hydroxy-7-methoxy-2-(4-methoxyphenyl)-

2,3-dihydrochromen-4-one

57

29

Zinc_18847037

Biochanin A

5,7-dihydroxy-3-(4-methoxyphenyl)

chromen-4-one

56

30

Zinc_120273

Galangin

3,5,7-trihydroxy-2-phenylchromen-

4-one

27. 28

31

Zinc_5999205

Glycitein

7-hydroxy-3-(4-hydroxyphenyl)-6-

methoxychromen-4-one

28. 49

32

Zinc_18847044

Prunetin

5-hydroxy-3-(4-hydroxyphenyl)-7-

methoxychromen-4-one

38

33

zinc_3871358

Acacetin\

5,7-dihydroxy-2-(4-methoxyphenyl)

chromen-4-one

28

34

Zinc_899093

Pectolinarigenin

5,7-dihydroxy-6-methoxy-2-(4-methoxyphenyl)

chromen-4-one

53

35

zinc_3871576

Apigenin

5,7-dihydroxy-2-(4-hydroxyphenyl)

chromen-4-one

28

36

Zinc_899915

Tectorigenin

5,7-dihydroxy-3-(4hydroxyphenyl)-6-

methoxychromen-4-one

50

37

Zinc_58116

Eriodictyol

2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-

2,3-dihydrochromen-4-one

27

38

Zinc_3872070

Chrysin

5,7-dihydroxy-2-phenylchromen-4-one

27. 28

39

Zinc_1785

Naringenin

5,7-dihydroxy-2-(4hydroxyphenyl)-2,

3dihydrochromen-4-one

28

40

Zinc_14806240

Dihydroechioidinin

hydroxy-2-(2-hydroxyphenyl)-7-

methoxy-2,3-dihydrochromen-4-one

40

41

Zinc_5999024

Naringenin 5-methyl ether

7-hydroxy-2-(4-hydroxyphenyl)-5-methoxy-

2,3-dihydro-4H-chromen-4-one

51

42

Zinc_57648

3,6-Dihydroxyflavone

3,6-dihydroxy-2-phenylchromen-

4-one

31

43

Zinc-18825330

Genistein

5,7-dihydroxy-3-(4 hydroxyphenyl)

chromen-4-one

27

44

zinc_33980812

Quercetin

2-(3,4-dihydroxyphenyl)-3,5,7-

trihydroxychromen-4-one

28

45

Zinc_73693

Pinocembrin

5,7-dihydroxy-2-phenyl-2,3-dihydro-

4H-chromen-4-one

28

46

Zinc_985403

Liquiritigenin

7-hydroxy-2-(4-hydroxyphenyl)-2,3-

dihydrochromen-4-one

28

47

Zinc_14806959

Baicalein

5,6,7-trihydroxy-2-phenylchromen-4-one

29

48

Zinc_2149675

5,7-Dihydroxyisoflavone

5,7-dihydroxy-3-phenylchromen-4-one

55

49

Zinc_57919

7-Hydroxyflavanone

7-hydroxy-2-phenyl-2,3-dihydrochromen-4-one

53

50

Zinc_18847034

Daidzein

7-hydroxy-3-(4-hydroxyphenyl)chromen-4-one

28

51

Zinc_1480711

Dihydrowogonin

5,7-dihydroxy-8-methoxy-2-phenyl-

2,3-dihydrochromen-4-one

44

These phytochemicals were screened in silico for their inhibitory activity against the selected enzyme molecules, comparing with 3-benzamido-2-hydroxybenzoic acid [fig. 3] which is known as inhibitor of KAS II.


Fig. 3: 3-benzamido-2-hydroxybenzoic acid which is known as inhibitor of KAS II

Protein-ligand docking

In this research, we use iGemdock (iGeneric Evolutionary Method Docking) program, which was used in various previous researches [63, 64, 65] and it is available for free [66, 67].

iGemdock v2.1

Docking software iGemdock was used to dock the protein of the enzyme (Kas II) with 50 flavonoids. iGemdock is an integrated virtual screening (VS) environment from preparations through post-screening analysis with pharmacological interactions iGemdock provides interactive interfaces to prepare both the binding site of the target enzyme and the screening compound library. Each compound in the library is then docked into the binding site by using the in-house docking tool iGemdock. Subsequently, iGemdock generates protein-compound interaction profiles of electrostatic (E), hydrogen-bonding (H), and Van der Waal’s (V) interactions. Based on these profiles and compound structures, iGemdock infers the pharmacological interactions and clusters the screening compounds for the post-screening analysis. Finally, iGemdock ranks and visualizes the screening compounds by combining the pharmacological interactions and energy-based scoring function of iGemdock

Rapid virtual screenings of the 50 ligand compounds were performed in the docking tooliGemdock. The docking consisted protocol "accurate docking" by setting population size of 800 is set with 80 generations and 10 solutions. After the completion of the docking, the post docking analysis was performed to find the docking pose and its energy values. The empirical scoring function of iGemdock was estimated uesing:

Energy = vdW+Hbond+Elec

Table 2 illustrates the result of the ten compounds based on the most favorable binding energy of flavonoids.

Table 2: The docking binding energy values results using iGEMDOCK

S. No.

Chemical name

Total binding energy

(kcal/mol)

Vanderwaals force

(kcal/mol)

H Bond

(kcal/mol)

Electrostatıc

1

Casticin

-112.5

-96.74

-15.76

0

2

Tangeritin

-101.55

-95.72

-5.83

0

3

Tamarixetin

-101.03

-87.03

-14

0

4

Malvidin

-100.92

-90.29

-10.63

0

5

Isorhamnetin

-98.17

-80.35

-17.81

0

6

Syringetin

-97.93

-88.55

-9.38

0

7

Petunidin

-97.65

-85.38

-13.37

0

8

Tricin

-97.32

-83.52

-14.81

0

9

Fisetin

-96.05

-81.88

-14.18

0

10

Herbacetin

-95.54

-81.72

-13.82

0

11

Benzoic acid, 3-(benzoylamino)-2-hydroxy

-92.76

-69.06

-15.38

-3.71


RESULTS AND DISCUSSION

In silico, docking studies were carried out using iGemdock v2.1. The results showed that all the selected flavonoids presented more favorable binding energy ranging from-112.5 kcal/mol to-80.9 kcal/mol when compared to that of the reference (-92.76 kcal/mol). Therefore, these molecular docking analyses could lead to further development of potent (Kas II) inhibitors for the prevention and treatment for diseases caused by Pseudomonas aeruginosa.

Table II summarizes results of the docking study based on binding energies. The energy, representing the best binding energy of inhibitors of this enzyme, was identified by the molecular docking procedure. In addition, fig. 4 illustrates the interactions of casticin with protein pocket, which has the most favourable binding energy and clarify the hydrogen bonding and Van der Waal’s interactions with the amino acids.


Fig. 4: Predicted docking pose of Casticin lie within the active site of the target protein (PDB ID-4JPF). Pink color represents the corresponding ligand molecule and green color represents the corresponding reference. Green and grey color represents the amino acids involved in hydrogen bonding and van der Waals interactions respectively

Table 3 shows pharmacological interactions and residues involved in the binding site for casticin. Then the pharmacological interactions are useful for understanding ligand binding mechanisms of a therapeutic target. These interactions are often inferred from a set of active compounds that were acquired by experiments.

Table 3: Pharmacological interactions and residues involved in the binding site

PDB ID

Predicted pharmacologica interactions

Casticin

4JPF

ARG-87-H-S

-9.1

GLY-84-H-M

-2.9

GLY-61-H-M

-2.5

GLU-61-V-M

-4.9

TYR-62-V-M

-5.5

TYR-62-V-S

-16.4

PHE-83-V-M

-8

PHE-83-V-S

-13

GLN-84-V-M

-11.2

GLN-84-V-S

-5.3

ARG-87-V-S

-16.3

Thegreen and grey color represents the amino acids involved in(H)hydrogen bonding and(V) van der Waals are interaction types M and S are Main chain and Side chain., In addition, the table 4 shows distance of hydrogen bond (Å) of some residues in the KAS II protein’s active site.


Table 4: The distance of hydrogen bond (Å) of some residues in the KAS II protein’s active site

Residues

Distance (Å)

ARG-87-H-S

1.99

GLY-84-H-M

2.17

GLY-61-H-M

2.04


Post screening analysis

The performed docking study against the KAS II receptor revealed that all flavonoid compounds identified in this study have a superior binding energy in comparison to the reference compound 3-benzamido-2-hydroxybenzoic acid. The analysis identified casticin (Zinc-6018556) as having the most favorable binding energy of-112.5 kcal/mol kcal/mol. Casticin is a methyoxylated flavonol, meaning the core flavonoid structure has methyl groups attached. Fig. 5 is shown the structure of casticin.


Fig. 5: The structure of casticin

The drug-receptor interactions and the fitness score of the compound suggest that this lead can be formulate as an antimicrobial activities drug against gram-negative Pseudomonas aeruginosa.

Lipinski’s rule of five

Lipinski et al. formulated the ’Rule of Five’ to relate likelihood of oral bioavailability which consists of four important properties (MW, log P, number of H bond donors/acceptors), each related to the number 5. The guidelines are based on data onto the literature for a large number of compounds, including all known drugs that correlate physical properties with oral bioavailability. The compounds are more likely to be membranes permeable and easily absorbed by the body if it matches the following criteria:

1. The molecular weight of less than 500 mg/mol

2. Has a high lipophilicity (log p less than 5)

3. Hydrogen bond donors less than 5

4. Hydrogen bond acceptor is less than 10

The rule describes molecular properties important for a drug's pharmacokinetics in the human body, including their Absorption, Distribution, Metabolism, and Excretion ("ADME").

The rule is important for drug development where a pharmacologically active lead structure is optimized step-wise for increased activity and selectivity, as well as drug-like properties as described by Lipinski's rules [68, 69, 70].

Compound classes that are substrates for biological transporters are exceptions to the rule. The molecular docking studies and Lipinski’s rules facilitate drug development avoiding expensive post clinical experiments.

Veber rule and Molar Refractivity

1-Veber Rule: In particular, compounds which meet only the two criteria of:

  1. 1-rotatable bond count>=10.
  2. 2-polar surface area (PSA) equal to or less than 140 Å.

Are predicted to have good oral bioavailability

2-Molar Refractivity: between (40-130) is used as measurement of the real volume of the molecule and it is also related to the forces, which govern the ligand-receptor interactions [70].

The 10 high ranked lead molecules were prioritized to follow Lipinski’s rules of five, veber rule and molar refractivity [71] based on the drug likeliness properties are listed in table 5.

Table 5: The Lipinski’s and Veber properties of the selected 10 ligands

Chemical name

Molecular Formula1

*M W2

g/mol

logP1 #

*X logP2

*HD 2

*HA2

*RB2

*(PSA)2Aο

*MR3

 

Value to be

500 >

5 >

 

5 >

10 >

=10 >

140 =>

40-130

Casticin

C 15 H 10 O 5

270.24

2.37

2.27

3

5

1

91

69.85

Tangeritin

C16H12O7

316.26

1.70

1.99

4

7

2

120

78.11

Tamarixetin

C16H12O7

316.26

1.67

1.99

4

7

2

120

78.11

Malvidin

C15H12O6

288.25

1.84

2.03

4

6

1

107

72.13

Isorhamnetin

C16H12O7

316.26

1.70

1.99

4

7

2

120

78.11

Syringetin

C 15 H 12 O 6

288.25

1.45

1.63

4

6

1

107

72.13

Petunidin

C21H22O9

254.24

2.49

2.56

2

4

1

71

67.97

Tricin

C16H14O6

302.28

1.93

1.94

3

6

2

96

76.93

Fisetin

C15H10O6

286.24

1.87

1.97

4

6

1

111

71.43

Herbacetin

C15H10O5

272.25

2.24

2.13

3

5

1

87

70.25

Benzoic acid, 3-(benzoylamino)-2-hydroxy

C15H10O4

254.24

2.72

2.94

2

4

1

70

67.67


1-calculated by ALOGPS 2.1 program http://www.vcclab.org/lab/alogps/start.html.

2-Calculated by www. zinc. docking. o, https://pubchem.ncbi.nlm.nih.gov/search/search.cgi

3-Calculated by ACD (Available Chemical Directory)

*PSA: Polar Surface Area,*MW: Molecular weight, *HD: H bond donor, *HA: H bond acceptor.

*RB: rotatable bonds. *MR: Molar refractivity

#Octanol/Water partition coefficient


CONCLUSION

The results of the present study clearly demonstrated that the in silico molecular docking studies of selected flavonoids with FabF enzyme exhibited binding interactions and warranted further studies needed for the development of potent FabF inhibitors for the treatment of Pseudomonas aeruginosa. These results clearly indicated that casticinhas similar binding sites and interactions with FabF compared to the reference.

These in silico studies are actually an added advantage to screen the FabF inhibition. Flavonoids may serve as useful leads in the development of clinically useful FabF inhibitors. Further, investigations on the above compound need in vitro and in vivo studies to develop potential chemical entities for the prevention and treatment of Pseudomonas aeruginosa infections.

ACKNOWLEDGMENT

The authors would like to thank all the management of Aleppo University and all the staff of Faculty of Pharmacy for assistance.

There is no known conflict of interest associated with the publication and there has been no significant financial support for this work that could have influenced its outcome.

CONFLICT OF INTERESTS

Declared None

REFERENCES

  1. Deza MA, Araujo M, Garrido MJ. Inactivation of escherichia coli, listeria monocytogenes, Pseudomonas aeruginosa and staphylococcus aureus on stainless steel and glass surfaces by neutral electrolyzed water. Lett Appl Microbiol 2005;40:341–6.
  2. Bell MT. The Use of Natural Products as Potential Anti-Pseudomonas Agents, Seton Hall University Dissertations and Theses. Spring; 2014. p. 5-17.
  3. MacDougall C, Harpe SE, Powell JP, Johnson CK, Edmond MB, Polk RE. Pseudomonas aeruginosa, Staphylococcus aureus, and fluoroquinolone use. Emerging Infect Dis 2005;11:1197–10.
  4. Fine MJ, Smith MA, Carson CA, Mutha SS, Sankey SS, Weissfeld LA, et al. Prognosis and outcomes of patients with community-acquired pneumonia. A Meta-Analysis. J Am Med Assoc 1996;275;134–41.
  5. Strateva T, Yordano D. Pseudomonas aeruginosa–a phenomenon of bacterial resistance. J Med Microbiol 2009; 58:1133–48.
  6. Arora D, Jindal N, Kumar R, Romit M. Emerging antibiotic resistance in pseudomonasa challenge. Int J Pharm Pharm Sci 2011;3:1488-91.
  7. Leeb M. A shot in the Arm. Nature 2004;431:892-5.
  8. Cronan JE Jr, Rock CO. In Escherichia coli and salmonella typhimurium: cellular and molecular biology. American Society for Microbiology Press: Washington, D. C; 1996.
  9. Lai CY, Cronan JE. β-ketoacyl-acyl carrier protein synthase III (FabH) is essential for bacterial fatty acid synthesis. J Biol Chem 2003;19:1494-03.
  10. Lu YJ, Zhang YM, Rock CO. Product diversity and regulation of type II fatty acid synthases. Biochem Cell Biol 2004;82:145-55.
  11. White SW, Zheng J, Zhang YM, Rock. The structural biology of type II fatty acid biosynthesis. Annu Rev Biochem 2005;74:791-31.
  12. Tung T, Schweizer HP. Characterization of pseudomonas aeruginosa enoyl-acyl carrier protein reductase (FabI): a target for the antimicrobial triclosan and its role in acylated homoserine lactone synthesis. J Bacteriol 1999;18:5489-97.
  13. Yuan Y, Sachdeva M, Leeds JA, Meredith TC. Fatty acid biosynthesis in pseudomonas aeruginosa is initiated by the fab Y Class of β-ketoacyl acyl carrier protein synthases. J Bacteriol 2012;194:5171-84.
  14. Zhang H, Machutta A, Tonge PJ. Fatty acid biosynthesis and oxidation, Stony Brook University. Stony Brook, NY, USA, Elsevier Ltd; 2010.
  15. Mengying H, Wu T, Pan S, Xu X. Antimicrobial mechanism of flavonoids against escherichia coli ATCC 25922 by model membrane study. Appl Surf Sci 2014;305:515–21.
  16. Céliz G, Daz M, Audisio MC. Antibacterial activity of naringin derivatives against pathogenic strains. J Appl Microbiol 2011;111:731–8.
  17. Jain A, Sinha P, Neetin S. Desai, Estimation of flavonoid, phenol content and antioxidant potential of indian screw tree (Helicteres Isora L.). Int J Pharm Sci Res 2014;5:1320-30.
  18. Kumar S, Pandey AK. Review article chemistry and biological activities of flavonoids hindawi publishing corporation. Sci World J 2013. doi.org/10.1155/2013/162750. [Article in Press]
  19. http://www.rcsb.org/pdb/home/home.do. [Last accessed on 01 Apr 2015].
  20. http://www.zinc.docking.orgkl. [Last accessed on 01 Apr 2015].
  21. Kumar S, Pandey AK. Chemistry and biological activities of flavonoids. Sci World J 2013. doi.org/10.1155/2013/162750. [Article in Press]
  22. Agyepong N, Agyare C, Adarkwa-Yiadom M, Gbedema SY. Phytochemical investigation and anti-microbial activity of clausena anisata (Willd). Hook. Afr J Tradit Complementary Altern Med 2014;11:200-9.
  23. Awolola GV, Koorbanally NA, Chenia H, Shode FO, Baijnath H. Antibacterial and anti-biofilm activity of flavonoids and triterpenes isolated from the extracts of ficus sansibarica warb. subsp. sansibarica (Moraceae) Extracts. Afr J Tradit Complementary Altern Med 2014;11:124-31.
  24. Hummelova J, Rondevaldova J, Balastikova A, Lapcik O, Kokoska L. The relationship between structure and in vitro antibacterial activity of selected isoflavones and their metabolites with special focus on antistaphylococcal effect of demethyltexasin. Biotechnol Appl Microbiol 2015;60:242-7.
  25. Morána A, Gutiérreza S, Martínez-Blancoab H, Ferreroab MA, Monteagudo-Meraa A, Rodríguez-Aparicioab LB. Non-toxic plant metabolites regulate staphylococcus viability and biofilm formation: a natural therapeutic strategy useful in the treatment and prevention of skin infections. Biofouling 2014;30:1175-82.
  26. Barbosa de Matos R, Braga-de-Souza S, Pena Seara Pitanga B, Amaral da Silva VD, Viana de Jesus EE, Morales Pinheiro A, et al. Flavonoids modulate the proliferation of neospora caninum in glial cell primary cultures. Korean J Parasitol 2014;52:613-9.
  27. L´azaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem 2009;9:31-59.
  28. Arima H, Ashida H, Danno G. Rutin enhanced antibacterial activities of flavonoids against bacillus cereus and salmonella enteritidis. Biosci Biotechnol Biochem 2002;66:1009-14.
  29. Shohaib T, Shafique M, Dhanya N, Madhu C, Divakar. Importance of flavonoides in therapeutics. Int J Nutr Pharmacol 2011;3:1-18.
  30. Novy P, Urban J, Leuner O, Vadlejch J, Kokoska L. In vitro synergistic effects of baicalin with oxytetracyclineand tetracycline against staphylococcus aureus. J Antimicrob Chemother 2011;66:1298-300.
  31. Urzua A, Modak B, Villarroel L, Torres R, Andrade L, Mendoza L, et al. External flavonoids from heliotropium megalanthum and H. huascoense (Boraginaceae) chemotaxonomic considerations. Bol Soc Chil Quim 2000:45:23-9.
  32. Anis S, Bhargava T, Upadhyay H. A review on phytotherapy by morus alba, review article. Int J Pharm Chem Sci 2012;1:1907-10.
  33. Liu J, Sridhar J, Foroozesh M. Cytochrome P450 Family 1 Inhibitors and structure-activity relationships. Molecules 2013;18:14470-95.
  34. Sultanova N, Makhmoor T, Abilov ZA, Parween Z, Omurkamzinova VB, Atta-ur-Rahman, et al. Antioxidant and antimicrobial activities of tamarix ramosissima. J Ethnopharmacol 2001;78:201-5.
  35. Fliniaux O, Corbin C, Ramsay A, Renouard S, Beejmohun V, Doussot J, et al. Microwave-assisted extraction of herbacetin diglucoside from flax (Linum usitatissimum L.) seed cakes and its quantification using an RP-HPLC-UV system. Molecules 2014;19:3025-37.
  36. Küçükboyaci N, Bilge Şener B. Two major flavonoids from the fruits of vitex agnus-castus L. Turk J Pharm Sci 2010;7:119-26.
  37. Aslam MS, Choudhary BA, Uzair M, Ijaz AS. The genus ranunculus: a phytochemical and ethnopharmacological review. Int J Pharm Pharm Sci 2012;4:15-22.
  38. Talib WH, Abu Zarga MH, Mahasneh AM. Antiproliferative, antimicrobial and apoptosis inducing effects of compounds isolated from inula viscosa. Molecules 2012;17:3291-03.
  39. Grecco SS, Gimenes L, Ferreira MJP, Romoff P, Favero OA, Zalewski CA, et al. Triterpenoids and phenolic derivatives from baccharis uncinella C. DC. (Asteraceae). Biochem Syst Ecol 2010;38:1234-7.
  40. Cushnie TP, Lambm AJ. Antimicrobial activity of flavonoids. Int J Antimicrob Agents 2005;26:343-56.
  41. Neeraja C, Hari Krishna P, Sudhakar Reddy C, Giri CC, Rao KV, Reddy VD. Distribution of andrographis species in different districts of andhra pradesh. Natl Acad Sci 2014;85:601-6.
  42. Urzua A, Modak B, Villarroel L, Torres R, Andrade L, Mendoza L, et al. External flavonoids from heliotropium megalanthum and H. Huascoense (Boraginaceae) chemotaxonomic considerations. Bol Soc Chil Quim 2000:45:23-9.
  43. Balouiri M, Sadiki M, Ouedrhiri W, Farah A, El Abed S, Koraichi SI. Antibacterial activity of extracts from salvia officinalis and rosmarinus officinalis obtained by sonication and maceration methods. Int J Pharm Pharm Sci 2014;6:167-70.
  44. Da Silva Filho AA, De Sousa JPB, Soares S. Antimicrobial activity of the extract and isolated compounds from baccharis dracunculifolia D. C. (Asteraceae). Z. Naturforsch C 2008;63:40-6.
  45. Lima CC, Lemos RPL, Conserva LM. Dilleniaceae family: an overview of its ethnomedicinal uses, biological and phytochemical profile. J Pharmacogn Phytochem 2014;3:181-04.
  46. Metsämuuronen S, Siren H. Antibacterial compounds in predominant trees in finland review. Metsämuuronen and siren. J Bioprocess Biotech 2014;4:1-13.
  47. Pal D, Mishra P, Sachan N, Ghosh AK. Biological activities and medicinal properties of cajanus cajan (L) Millsp. J Adv Pharm Technol Res 2011;2:207–14.
  48. Wiart C. Goniothalamus species: a source of drugs for the treatment of cancers and bacterial infections. J Evidence-Based Complementary Altern Med 2007;4:299–11.
  49. Borik RM. Isolation and structural characterization of a steroidal antimicrobial agent from clerodendrum baronianum. World J Chem 2013;8:48-54.
  50. Brand-Garnys EE, Denzer H, Meijer H, Brand HM. Flavonoids: a review for cosmetic application. part I. J Appl Cosmetol 2007;25:93-109.
  51. Joung DK, Mun SH, Lee KS. The antibacterial assay of tectorigenin with detergents or ATPase inhibitors against methicillin-resistant staphylococcus aureus. Hindawi 2014. doi.org/10.1155/2014/716509. [Article in Press]
  52. Hammami S, Jannet HB, Bergaoui A, Ciavatta L, Cimino G, Mighri Z. Isolation and structure elucidation of a Flavanone, a Flavanone glycoside and vomifoliol from echiochilon fruticosum growing in tunisia. Molecules 2004;9:602-8.
  53. Bastos MLA, Lima MRF, Conserva LM, Andrade VS, Rocha EM, Lemos RP. Studies on the antimicrobial activity and brine shrimp toxicity of zeyheria tuberculosa (Vell.) Bur. (Bignoniaceae) extracts and their main constituents. Ann Clin Microbiol Antimicrob 2009;8:16.
  54. Singh MK, Khare G, Iyer SK, Sharwan G, Tripathi DK. Clerodendrum serratum: a clinical approach. Int J Pharm Sci 2012;02:11-5.
  55. Filho AAO, Fernandes HMB, Sousa JP, Maia GLA, Barbosa-Filho JM, Lima EO, Oliveira TL. Actividad antibacterial del flavonoide 5.7.4’-Trimetoxiflavona aislada de Prexelis clematides R. M. King and Robinson. Bol Latinoam Caribe Plant Med Aromat 2013;12:400-4.
  56. Gargala G, Baishanbo A, FavennecL, François A, Ballet J, Rossignol JF. Inhibitory activities of epidermal growth factor receptor tyrosine kinase-targeted dihydroxyisoflavone and trihydroxydeoxybenzoin derivatives on sarcocystis neurona, Neospora caninum, and cryptosporidium parvum development. Antimicrob Agents Chemother 2005;49:4628–34.
  57. Liu G, Liang JC, Wang XL, Li ZH, Wang W, Guo N, et al. In vitro synergy of biochanin a and ciprofloxacin against clinical isolates of staphylococcus aureus. Molecules 2011;16:6656-66.
  58. Johari SA, Kiong LS, Mohtar M, Isa MM, Man S, Mustafa S, et al. Efflux inhibitory activity of flavonoids from chromolaena odorata against selected methicillin resistant staphylococcus aureus (MRSA) isolates. Afr J Microbiol Res 2012;6:5631-5.
  59. Azlan A, Younis L, Mahmud NH, Dardiri NA. Mechanisms of action of andrographis paniculata as anti-atherosclerotic agent. Eur Int J Sci 2013;2:91-6.
  60. Bell MT. The Use of Natural Products as Potential Anti-Pseudomonas Agents, Seton Hall University Dissertations and Theses. Spring; 2014. p. 5-17.
  61. Wright HT, Reynolds KA. Antibacterial targets in fatty acid biosynthesis. Curr Opin Microbiol 2007;10:447–53.
  62. Tapas AR, Sakarka DM, Kakde RB. Flavonoids as nutraceuticals. Trop J Pharm Res 2008;7:1089-99.
  63. Gnanslin Sheeba D, Subha V, Suseela Gomathi K, Citarasu T. Virtual docking studies of flavonoid compounds against cell wall proteins of mycobacterium tuberculosis. Asian J Pharm Res Dev 2013;1:88-97.
  64. Bugata BK, Kaladhar DSVGK. QSAR and docking studies of synthesized diarylsulfonylurea chalcone hybrids as anti-inflammatory agents. Int J Pharm Sci Rev Res 2014;24:144-9.
  65. Chen CC, Er TK, Liu YY, Hwang JK, Barrio MJ, Rodrigo M, et al. Computational analysis of KRAS mutations: implications for different effects on the KRAS p. G12D and p. G13D mutations. PLoS One 2013;8:e55793.
  66. Kai CH, Yen FC, Shen RL, Jinn MY. IGEMDOCK: a graphical environment of enhancing GEMDOCK using pharmacological interactions and postscreening analysis. BMC Bioinf 2011;12(Suppl 1):S33.
  67. Yang JM. Graphical-Automatic Drug Design System for Docking, Screening and Post-Analysis. Department of Biological Science and Technology and Institute of Bioinformatics National Chiao Tung University; 2008. p. 1-69.
  68. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Delivery Rev 2001;46:3–26.
  69. Thomas G. Medicinal chemistry. 2nd Edition. University of Portsmouth; 2007. p. 9-10.
  70. Kerns EH, Di L. Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization. Academic Press is an imprint of Elsevier; 2008. p. 6-120.
  71. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Delivery Rev 2001;46:3–26.
  72. Zhang YM, White SW, Rock CO. Inhibiting bacterial fatty acid synthesis. J Biol Chem 2006;281:17541-4.