Next Article in Journal
Eosinophilia as Monitoring Parameter for Chronic Graft-versus-Host Disease and Vitamin D Metabolism as Monitoring Parameter for Increased Infection Rates in Very Long-Term Survivors of Allogeneic Stem Cell Transplantation—A Prospective Clinical Study
Previous Article in Journal
Biological Reference Intervals for 17α-Hydroxyprogesterone Immunoreactive Trypsinogen, and Biotinidase in Indian Newborns
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytochemical Composition, In Silico Molecular Docking Analysis and Antibacterial Activity of Lawsonia inermis Linn Leaves Extracts against Extended Spectrum Beta-Lactamases-Producing Strains of Klebsiella pneumoniae

by
Adam Mustapha
1,†,
Ahmed Nouri AlSharksi
2,
Ukpai A. Eze
3,*,†,‡,
Rahma Kudla Samaila
1,
Boniface Nwofoke Ukwah
4,
Arinze Favour Anyiam
5,6,
Shivanthi Samarasinghe
3 and
Musa Adamu Ibrahim
7
1
Department of Microbiology, Faculty of Life Sciences, University of Maiduguri, Maiduguri PMB 1069, BO, Nigeria
2
Department of Microbiology, Faculty of Medicine, Misurata University, Misrata 93FH+66F, Libya
3
Leicester School of Allied Health Sciences, Faculty of Health of Life Sciences, De Montfort University, Leicester LE1 9BH, UK
4
Department of Medical Laboratory Sciences, College of Health Sceinces, Ebonyi State University, Abakaliki PMB 053, EB, Nigeria
5
Department of Medical Laboratory Science, School of Basic Medical and Health Sciences, Igbinedion University, Okada 302110, ED, Nigeria
6
Department of Medical Laboratory Science, Faculty of Applied Health Sciences, Edo State University, Uzairue 200099, ED, Nigeria
7
Department of Biology, Faculty of Life Sciences, University of Maiduguri, Maiduguri PMB 1069, BO, Nigeria
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current address: Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH2 1BR, UK.
Submission received: 27 June 2024 / Revised: 16 August 2024 / Accepted: 19 August 2024 / Published: 26 August 2024

Abstract

:
Klebsiella pneumoniae is an opportunistic Gram-negative bacterium in the Enterobacteriaceae family associated with a wide range of diseases, such as pneumonia, bloodstream infections, meningitis and urinary tract infections. Infections caused by drug-resistant strains of Klebsiella pneumoniae pose a significant threat to the effectiveness of conventional antibiotics. Hence, this has led to the need to explore alternative antimicrobial therapies, especially natural products derived from plant sources. This study assessed the phytochemical composition and antibacterial properties and performed a molecular docking analysis of Henna leaves (Lawsonia inermis L.) extracts on strains of Klebsiella pneumoniae. Crude ethanol and methanol extracts of L. inermis L. were prepared at different concentrations (25, 50, 75 and 100 mg/mL) and tested on extended spectrum beta-lactamases (ESBLs)-producing strains of Klebsiella pneumoniae. Phytocompounds were identified using gas chromatography–mass spectrometry (GC-MS) and further subjected to virtual ligands screening with DataWarrior (v05.02.01) and a molecular docking analysis using AutoDock4.2 (v4.2.6). The active compounds of L. inermis L. were determined by the docking analysis, including phytochemical, physicochemical, pharmacokinetics and docking score. The GC-MS analysis identified 27 phytoconstituents, including ethyl acetate, sclareol, 2-[1,2-dihydroxyethyl]-9-[β-d-ribofuranosyl] hypoxanthine, α-bisabolol and 2-Isopropyl-5-methylcyclohexyl 3-(1-(4-chlorophenyl)-3-oxobutyl)-coumarin-4-yl carbonate. The 27 compounds were then screened for their physicochemical and pharmacokinetic properties. The results revealed that the methanol extracts at 100 mg/mL showed significantly higher (p < 0.05) zones of inhibition (13.7 ± 1.2 mm), while the ethanol extracts at 50 mg/mL were significantly lower (6.3 ± 0.6 mm) compared to all the other treatments. The docking analysis revealed that out of the 27 compounds identified, only twelve (12) compounds have a drug-likeness activity. The 12 compounds were further subjected to docking analysis to determine the binding energies with the CTX-M protein of Klebsiella pneumoniae. Only one compound [CID_440869; (2-[1,2-dihydroxyethyl]-9-[β-d-ribofuranosyl] hypoxanthine)] had the best binding energy of −9.76 kcal/mol; hence, it can be considered a potentially suitable treatment for infections caused by ESBLs-producing strains of Klebsiella pneumoniae. This study has demonstrated that L. inermis L. extracts have antibacterial effects. Further research could explore the potential antimicrobial applications of L. inermis L. extracts to many bacterial strains.

1. Introduction

The rising problem of antibiotic resistance in microorganisms poses a threat to global public health [1]. This aligns with the projections made by De Kraker et al. [2], estimating that antibiotic-resistant organisms could cause 10 million deaths annually by 2050 if strategies are not put in place to curtail the rising trend of antimicrobial resistance. The threat of antibiotic resistance is beyond clinical and public health; it leads to huge economic loss due to long hospital stays, reduced productivity and overworked healthcare systems [2]. A recent worldwide analysis of antimicrobial resistance-associated deaths highlights Sub-Saharan Africa as the most affected region, with Escherichia coli, Staphylococcus aureus, Klebsiella pneumoniae, Streptococcus pneumoniae, Acinetobacter baumannii and Pseudomonas aeruginosa topping the list of bacterial-associated deaths due to antimicrobial resistance [3]. Antibiotics have brought rays of hope to modern medicine in the treatment of wide ranges of infections; however, the emergence of antibiotic resistance undermines this effort [4,5]. Despite the gravity of this threat, common available antibiotics have proven ineffective in eradicating these infections [4,5].
For decades, plants have been a historical source of medicinal remedies garnering global attention as alternatives to conventional drugs, particularly during the COVID-19 pandemic [4,6,7]. In particular, the post-COVID-19 pandemic period has further exacerbated the problem of antibiotic resistance due to the increased use of antimicrobials in an attempt to reduce the gravity of COVID-19 clinical outcomes on a global scale [8,9]. The use of natural products as a source of bioactive compounds for their antibacterial activity is well documented in the treatment of multidrug-resistant pathogens and this has gained attention in recent years [10,11,12]. For instance, Arum maculatum exhibited antibacterial activity against Staphylococcus aureus, Listeria monocytogenes, Escherichia coli and Pseudomonas aeruginosa [10]. Another plant with antibacterial activity was reported by Wasihul et al. [11], and they demonstrated that Calpurnia aurea contained phytochemical components, such as alkaloid, tannins, flavonoid and saponins, which suggested their antibacterial activity against both Gram-positive and Gram-negative bacteria. Similarly, a phytochemical screening of Pulicaria spp revealed secondary metabolites, and the extract revealed significant activity against bacterial strains [12]. Many studies have reported the presence of bioactive phytochemicals as secondary metabolites, including alkaloids, amino acids, flavonoids, phenylpropanoids, steroids, volatile oils, glycosides, terpenes, anthraquinones and other compounds [4,7,13]. Medicinal plants demonstrate remarkable antimicrobial activities due to different phytochemical components they possess [14]. This gives the basis for the application of plants and herbs for medicinal purposes [15,16]. This assertion is backed by the World Health Organization’s report that more than 80% of the world’s population use medicinal plants for their basic health care [17].
Lawsonia inermis Linn., also known as henna in many parts of the world, is a shrub classified in the family Lythraceae. Lawsonia inermis L. is found in many regions but is considered native to Africa, Middle East and Asia. It is widely found in Africa, especially East and West African countries, and in certain regions of South Africa [18]. It is a plant with many names that are largely region-specific: Arabic: henna; Bengali: Mendi, mehedi; English: Egyptian privet, henna, Jamaica mignonette, mignonette tree; French: henné; German: Hennastrauch; Hindi: mehndi; Indonesian: inai, pakar kuku; Portuguese: hésia, hena, alfeneiro; Spanish: alcana, alheña; Swedish: henna; Vietnamese: nhuôm móng taylâ mòn; and in Nigeria, particularly northern Nigeria, where it is mostly used, it goes by the name Lalle [18].
In addition to its cosmetic uses, this plant species is known to contain many bioactive compounds with therapeutic potential in the treatment of infections [15]. In a recent study, Fatahi et al. [16] demonstrated that the different sub-types of Lawsonia inermis L. (Shahdad, Rudbar and Ghale-e-ganj) exhibited antibacterial and anti-trichomonas effects, particularly inhibiting the growth of Streptococcus agalactiae, Pseudomonas aeruginosa and Trichomonas vaginalis in vitro. Several studies have shown that Lawsonia inermis L. contains flavonoids, phenolic compounds, proteins, saponins, terpenoids, alkaloids, xanthones, resin, quinones, coumarins and tannins, and these chemical components have been suggested to mediate their medicinal activities [18,19,20].
K. pneumoniae is a Gram-negative bacterium of the Enterobacteriaceae family and causes infections such as pneumonia, meningitis, wound infections and surgical site infections, urinary tract infections, bloodstream-associated infections and respiratory tract infections [21]. Most strains of K. pneumoniae produce extended-spectrum beta-lactamases (ESBLs), which confer resistance to most beta-lactam antibiotics, including penicillin, extended-spectrum cephalosporins (cefotaxime, ceftriaxone, ceftazidime and cefepime) and monobactams (aztreonam) [22,23]. The most common families of ESBLs that have been identified globally include CTX-M, SHV, OXA and TEM [22,23,24]. However, the majority of the ESBLs produced by K. pneumoniae belong to the CTX-M enzymes, which have been recognized as critical public health threats, both in hospital settings and in the community [22,23,24,25,26]. With the emergence of multidrug resistance in K. pneumoniae and the abundance of medicinal plants, there is a need for effective screening of new compounds and ultimately extraction and characterization of specific bioactive agents [27,28]. Gas chromatography–mass spectrometry (GC–MS) has been a mainstay in the identification of functional groups and specific bioactive compounds present in medicinal plants. GC-MS is considered a rapid and effective technique to profile novel phyto-constituents in medicinal plants when compared with the standards deposited in the National Institute of Standards and Technology Mass Spectra database (NIST) [29]. In addition, a computer-aided approach is employed to elucidate the medicinal informatics of plants via a virtual screening method [29]. This approach has been used in silico to predict pharmacokinetic, pharmacological and toxicological profiles of medicinal plants, and it is considered a fast and cost-effective method of testing potential drug candidates of bioactive compounds against target proteins [27,28,29,30,31,32,33]. In a general context, in silico molecular docking can be used to evaluate the interaction between proteins and ligands to simulate their binding ability while elucidating the structure. Some studies on the antibacterial activity of Lawsonia inermis L. against Klebsiella pneumoniae have been reported [34,35,36,37,38]. For example, the antibacterial effects of different concentrations of the chloroform henna extracts (CHE) were tested against S. aureus (100 mg/mL) and K. pneumoniae (200 mg/mL) in vitro, and the results revealed higher activity than the standard antibiotic control, ciprofloxacin [34]. Additionally, Arun et al. [35] demonstrated high flavonoid contents of L. inermis L., and the methanolic extract revealed antibacterial activity against different bacterial strains. Similarly, Pasadi et al. [36] demonstrated that aqueous extracts from three henna ecotypes exhibited antibacterial effects in a dose-dependent manner, with K. pneumoniae and B. cereus showing higher resistance. Furthermore, various studies have demonstrated the antibacterial activity of L. Inermis L. against Klebsiella pneumoniae [37,38,39].
Although the antibacterial activity of henna has been investigated, multi-approaches to investigate the in vitro antibacterial activity and molecular docking analysis of the plant against multidrug-resistant bacterial pathogens have not been reported to the best of our knowledge. Therefore, the current study investigated (i) the in vitro antibacterial activity of L. inermis L. against Klebsiella pneumoniae; (ii) the bioactive compounds in L. inermis L. through the GC-MS technique; and (iii) the in silico molecular docking analysis of potential compound from L. inermis L. against the CTX-M protein of ESBLs-producing Klebsiella pneumoniae strains.

2. Material and Methods

2.1. Bacterial Isolates

Klebsiella pneumoniae isolates were obtained from the Department of Microbiology, Faculty of Life Sciences, University of Maiduguri, Nigeria. Standard microbiological and biochemical methods were used to phenotypically identify and characterize the Klebsiella pneumoniae isolates as described by Cheesbrough [40]. Furthermore, a pure culture of the isolates was maintained using streak plating on MacConkey agar plates and incubating at 37 °C for 24 h.

2.2. Collection and Preparation of Plant Sample

Fresh leaves of Lawsonia inermis L. were collected during the morning hours from the mother tree and taken to the Department of Botany, University of Maiduguri, Nigeria, for proper identification and authentication. After authentication, the leaves were taken to the Microbiology Department of the same university for processing. The leaves were washed thoroughly using distilled water and allowed to air-dry for 3–5 days under a controlled environment in the laboratory at room temperature. After drying, the leaves were pulverized with a clean grinder and then sieved to obtain a fine powder of Lawsonia inermis L. Approximately 200 g of the dried powder was extracted with 500 mL of ethanol and methanol, respectively, through a soxlet extractor, and the extracts were filtered using Whatman No. 1 filter paper. The solvents were vaporized using a rotary evaporator, and the crude extracts were stored at 4 °C until further assay. Samples of the Lawsonia inermis L. leaves used in this study have been deposited in the Department of Botany, University of Maiduguri, Nigeria.

2.3. Determination of the Plant Extracts’ Yield and Phytochemical Screening

The leaves’ extract yield (%) were determined using the formular by Nabi et al. [41]:
Yield (%W/W) = W1 × 100/W2
where W1 represents the dry weight of the extract after solvent evaporation and W2 represents the weight of the dried leaf powder.
The methanol and ethanol leaf extracts of Lawsonia inermis L. were evaluated for the presence of secondary metabolites, such as tannins, saponins, flavonoids and anthraquinones, following the standard protocols as described by Gul et al. [42].

2.4. Analysis of the Phytochemical Composition Using Gas Chromatography–Mass Spectrometry (GC-MS)

The procedure employed was as previously reported by Idris et. al. [39]. Briefly, 10 g of the dried crude methanolic extract of Lawsonia inermis L. was introduced into a centrifuge tube containing 10 mL of methanol and then mixed properly by vortexing for 2 min, after which it was centrifuged for 10 min at 3000 rpm. After centrifugation, a clear supernatant was taken and dispensed into a TSP micro vial, which was then subjected to GC-MS analysis. The GC analysis was carried out first by injecting 1 µL of the supernatant into Agilent GC (7890 B), equipped with a 30 m × 250 µm × 0.25 µm column, coupled with Agilent Mass Selective Detector (MSD) 5977 A technologies. The GC-MS has a carrier containing helium gas, which was set at a flow rate of 1 mL/min. Before the analysis was performed, the GC oven was kept at a temperature of 70 °C for about 3 min and then elevated at 10 °C/min to 280 °C. The temperature was held for 9 min. The equilibration time was set at 0.5 min, the MSD transfer line temperature was 250 °C, the MS source temperature was 230 °C and the MS quad temperature was 150 °C. The chemical compounds in the methanol extracts of Lawsonia inermis L. were detected and identified using the retention time produced by GC. The mass spectrum was then matched with the mass spectrum data available in the database of the National Institute of Standards and Technology. The percentage composition of each sample constituent was expressed as a percentage by peak area.

2.5. Phenotypic Detection of Extended-Spectrum β-Lactamases (ESBLs) Production in K. pneumoniae Isolates

The screening for ESBLs production in all the K. pneumoniae isolates was performed using the modified double-disc synergy (MDDS) test, as described by Wakil et al. [43]. Using a sterile pipette tip, colonies of the ESBLs-producing Klebsiella pneumoniae were picked and suspended in 2 mL of sterile phosphate buffered saline. The suspension was thoroughly mixed and then compared to a 0.5 MacFarland standard to match the level of turbidity using a piece of black paper. Afterwards, 100 μL of the bacterial suspension was pipetted onto the center of the Mueller–Hinton agar (MHA) plates and spread aseptically onto the surface of the Mueller–Hinton agar using a sterile glass spreader. Thereafter, ceftazidime (CAZ 30 μg), cefotaxime (CTX 30 μg) and ceftriaxone (CRO 30 μg) were placed on each of the isolates inoculated on the Mueller–Hinton agar plates and incubated at 37 °C for 24 h. The diameter of the zones of inhibition around each of the discs was measured, and a zone of inhibition diameter of 5 mm or higher signified the presence ESBLs production [43].
The isolates with an indication of the presence of ESBLs production in the MDDS test were further confirmed by placing ceftazidime (CAZ 30 μg), cefotaxime (CTX 30 μg) and ceftriaxone (CRO 30 μg) on each side of an Augmentin (AMC 30 μg) disc at a distance of 15 mm from the middle of the inoculated Mueller–Hinton agar plates, and the plates were incubated overnight at 37 °C for 24 h. The zone of inhibitions towards the Augmentin (AMC 30 μg) is a confirmation of ESBL production by the tested isolates [43,44].

2.6. Evaluation of the Antibacterial Activity of Both Methanol and Ethanol Extracts of Lawsonia inermis L.

The agar well diffusion method was employed to determine the antibacterial activities of Lawsonia inermis L. extracts. Using a sterile pipette tip, colonies of the ESBLs-producing Klebsiella pneumoniae were picked and suspended in 2 mL of sterile phosphate buffered saline. The suspension was thoroughly mixed and then compared to a 0.5 MacFarland standard to match the level of turbidity using a piece of black paper. Afterwards, 100 μL of the bacterial suspension was pipetted onto the center of the Mueller–Hinton agar (MHA) plates and spread aseptically onto the surface of Mueller–Hinton agar using a sterile glass spreader. Then, wells 10 mm in diameter were cut from the inoculated agar with separate sterile cork-borers. The wells were then filled with different concentrations (25, 50, 75 and 100 mg/mL) of the methanol and ethanol extracts of Lawsonia inermis L., respectively. Methanol and ethanol were used as controls. These plates were incubated at 37 °C for 24 h, followed by the measurement of the zones of inhibition in millimeters using a Vernier caliper. Each antibiotic underwent testing in triplicates over four days, and the averages were calculated (mean ± standard deviation).

2.7. Preparation of the Crystal of CTX-M Target Protein

The protein of Klebsiella pneumoniae (CTX-M) was complexed with GDP and 9PC (PDB ID: 4DXD), which was obtained from the Protein Data Bank (PDB). The ligand within the bound structure of CTX-M was removed and then cleaned properly. Missing factors, which include atoms, residues, loops and side chains, were verified properly and then inserted. The Chimera, Swiss PDB Viewer and Chiron energy minimization and refinement tool (version 1.0, Swiss National Science Foundation, Wildhainweg, Switzerland) were then used to eliminate all the water molecules, especially those not closer to the binding site of the substrate. In addition, non-proteinate residues were removed through the optimization of the structure and minimization of energy [45,46,47].

2.8. Physicochemical Analysis

All the compounds obtained from GC-MS analysis were screened based on their physicochemical properties (molecular weight, logarithms of partial coefficient, number of hydrogen-bond donors (HBAs) and number of hydrogen-bond acceptors (HBDs)) using the DataWarrior (v05.02.01) program [48,49]. All the compounds with suitable physicochemical properties were selected for additional evaluation.

2.9. Pharmacokinetic Analysis

The identified compounds that have good binding energies and physicochemical analysis were then used to evaluate the compounds on the basis of their pharmacokinetics properties, which include absorption, distribution, metabolism and excretion (ADMET), using the AdmetSAR version 3.0, “http://lmmd.ecust.edu.cn/admetsar3/ (assessed on 12 April 2024)” tool as described by Cheng et al. [50], the DataWarrior (v05.02.01) program by Sander et al. [48] and SwissADME (version 2.3.0) by Daina et al. [49]. Other properties identified include mutagenicity, tumorigenicity, reproductive toxicity and irritant.

2.10. Molecular Docking Analysis

A molecular docking analysis was carried out in order to determine the conformation of binding between the CTX-M protein and the ligand forming a protein–ligand complex with the aid of AutoDock4.2 (v4.2.6), which was employed by Morris et al. [51]. The binding confirmation of the complex reveals the binding energy of CTX-M and the selected ligands. The binding energy of the protein–ligand complex was calculated using the formula by Hariono et al. [52]:
∆Gbind = ∆Gvdw + ∆Ghbond + ∆Gelect + ∆Gconform + ∆Gtor + ∆Gsol
where ∆Gbind = estimated free binding energy;
∆Gvdw = sum of van der Waals energy;
∆Ghbond = sum of hydrogen bond and desolvation energy;
∆Gelect = sum of electrostatic energy;
∆Gconform = sum of final total internal energy;
∆Gtor = sum of torsional free energy;
∆Gsol = sum unbound system energy.
The binding energies and residues were recorded appropriately.

3. Results

3.1. Percentage Yield and Phytochemical Screening

The percentage yield (%w/w) of the dried leaf extracts was 55% for methanol and 48% for the ethanol extracts. The results of the preliminary phytochemical screening of both the methanolic and ethanolic extracts are presented in Table 1.

3.2. Compounds Identified from Lawsonia inermis L. Using Gas Chromatography-Mass Spectroscopy (GC-MS)

A gas chromatography–mass spectrometry analysis was carried out on the methanolic extract of the leaves of L. inermis L. in order to identify the phytochemical parameters present in the leaves of L. inermis L. The analysis revealed the different constituents of the phytochemicals, including their compound names, chemical formula, peak value and retention time. The spectrum obtained from the GC-MS analysis is shown in Figure 1. The analysis carried out on the methanolic extract of L. inermis L. revealed the existence of twenty-seven (27) compounds. The compound with PubChem ID CID_6590 (C4H8O2) has the lowest molecular weight, which is 88, while CID_ 53178 (C30H33ClO6) has the highest molecular weight of 524 (Table 2).

3.3. Antibacterial Activity of the Methanol and Ethanol Extracts of Lawsonia inermis L.

The antibacterial activity of both the methanol and ethanol extracts of L. inermis L. of different concentrations (25, 50, 75 and 100 mg/mL) were tested on isolates of multidrug-resistant Klebsiella pneumoniae isolated from clinical samples of urine and wounds and assessed for the presence and absence of an inhibition zone. The antibacterial activity was assessed by measuring the inhibition zone diameter. The methanol extract of L. inermis L. had the highest inhibition zone of 13.00 ± 1.2 mm at a concentration of 100 mg/mL, whereas the lowest inhibition zone of 7.3 ± 0.6 mm was seen at a concentration of 75 mg/mL. The ethanol extract of L. inermis L. revealed that a concentration of 25 mg/mL had the highest concentration of 11.00 ± 0.00 mm, followed by 100 mg/mL, which has 10.00 ± 1.0 mm. The lowest inhibition zone was seen at the concentration of 50 mg/mL, which had an inhibition zone of 6.00 ± 0.6 mm. These results are shown in Table 3.

3.4. Physiochemical Analysis of Compounds Obtained from Lawsonia inermis L.

The physicochemical analysis revealed that all the compounds are in agreement with the five rules of Lipinski and Egan, with the exception of CID_440869 and CID_537118. CID_440869 has 11 hydrogen bond acceptors (HBA ≤ 10) and 6 hydrogen bond donors (HBD ≤ 5), despite having a molecular weight of 328.280 Da, whereas CID_537118 has a weight of 525.039 Da, which is above the required molecular weight of 500 Da. In addition, CID_537118 has a logarithm of the partial coefficient of 7.3414 as opposed to the required value of (≤5) (Table 4). Therefore, apart from CID_440869 and CID_537118, all the other compounds have drug-like characteristics (Table 4).

3.5. Pharmacokinetic Analysis of the Compounds Detected from Lawsonia inermis L.

The pharmacokinetic properties of L. inermis L. were determined using parameters such as mutagenicity, tumorigenicity, reproductive toxicity and irritant. The pharmacokinetic properties of a drug are used to determine the effectiveness and impact of the drug. In this study, the following compounds failed pharmacokinetic analysis due to varying mutagenic properties: CID_225038, CID_76029, CID_5365831, CID_551300, CID_225038, CID_76029, CID_5365831 and CID_551300.
Compound CID_11996452 was the only compound that had evidence of tumorigenicity, while CID_8842 and CID_537118 had high reproductive toxicity. Based on irritant, CID_6590, CID_536980, CID_8180, CID_11996452, CID_8842, CID_5363274, CID_5365831, CID_319068771, CID_296248 and CID_537118 have a high irritant ability, while CID_225038, 534592, 163263 and CID_551300 have low irritant capacity (Table 5). Furthermore, CID_8842 did not meet the pharmacokinetic properties of an ideal compound due to its high toxicity and irritability.

3.6. Docking Scores and Residues Involved in H-Bond Formation

A molecular docking analysis was carried out on the twelve compounds to evaluate their binding energies with the Klebsiella protein (CTX-M). The molecular docking revealed free binding energies ranging from −3.62 kcal/mol to −9.76 kcal/mol. Five of the compounds, CID_5363192, CID_440869, CID_5363411, CID_5283028 and CID_535324, produced residues that are included in hydrogen bonding. They include Leu200, Pro201, Gln197, Val206, Glu262, Lys257, Arg229, Trp204 and Gly199 (Table 6).

4. Discussion

Medicinal plants are rich sources of secondary metabolites that are responsible for antibacterial activity against many pathogens and exhibit less adverse effects [4,7]. However, there is little or no scientific validation or documentation on the application of these medicinal plants. In the current study, the methanol extract of the leaves of L. inermis L. was used for the detection of its bioactive compounds. A total of twenty-seven (27) phytocompounds were detected with the aid of the GC-MS on the methanol extract of L. inermis L. (Table 2). The analysis shows a wide range of potential bioactive agents, which could serve as candidates to inhibit ESBL-producing strains of K. pneumoniae. The 27 phytocompounds reported in this study suggest that some of the compounds have antibacterial activity (Table 3, Table 4 and Table 6).
The GC-MS analysis of the L. inermis L. identified 27 phytoconstituents, including ethyl acetate, sclareol, 2-[1,2-Dihydroxyethyl]-9-[β-d-ribofuranosyl] hypoxanthine, alpha-bisabolol and 2-Isopropyl-5-methylcyclohexyl 3-(1-(4-chlorophenyl)-3-oxobutyl)-coumarin-4-yl carbonate. These phyto-compounds are likely to contribute to the antibacterial activity of the plants. Previous studies reported some of the phyto-compounds in this study to have antibacterial effect [15,16,18,19]. For instance, Jeyaseelan et al. [53] demonstrated that both ethyl acetate and ethanol extracts of the fruits, flowers and leaves of Lawsonia inermis L. generally contained flavonoids and had antimicrobial activity against E. coli, P. aeruginosa, B. subtilis and S. aureus. This correlated with the findings in the current study, which also demonstrate that the ethanol extract of Lawsonia inermis L. leaves had antibacterial activity against ESBL-producing K. pneumoniae. In a recent study, Popova et al. [54] evaluated the antimicrobial activities of sclareol, and it was demonstrated that sclareol had significant antimicrobial effects against Bacillus cereus ATCC 11778, Escherichia coli ATCC 8739, Salmonella abony ATCC 6017, Staphylococcus aureus ATCC 6538, Proteus mirabilis ATCC 14153 and Proteus vulgaris ATCC 13315, with zone diameters of inhibition ranging from 9.5 ± 0.10 mm to 14.2 ± 0.06 mm [54]. In the same study, Popova et al. [54] reported that sclareol had strong antimicrobial effects towards various fungal isolates, including Candida albicans АТСС 10231, C. glabrata ATCC 90030, C. parapsilosis clinical isolate and C. tropicalis NBIMCC 23, with their zones of inhibition diameters similar to those of the antifungal agent fluconazole [54]. A checkerboard analysis of combinations of sclareol with curcumin and sclareol with eugenol showed strong synergistic antimicrobial activities against C. albicans, C. glabrata and Aspergillus fumigatus, with MICs reduced by up to four- and eight-fold, respectively [55]. In a different study, synergism assays of sclareol with clindamycin were performed by checkerboard assay, and the results indicated that sclareol had a synergistic effect with clindamycin towards methicillin-resistant Staphylococcus aureus [56]. Furthermore, sclareol has been shown to exhibit antiviral activity, especially in Ebola virus, where it was found to block the Ebola viral fusion process, thereby interfering with virus entry into host cells [57].
Alpha-bisabolol, also known as levomenol, is a sesquiterpenoid that was first isolated from German chamomile but is also found in various medicinal plants [58]. Rodrigues et al. [59] reported that alpha-bisabolol exhibited significant antimicrobial activity against E. coli, S. aureus, Candida albicans, C. krusei and C. tropicalis, and α-bisabolol in combination with aminoglycosides and beta-lactams had strong synergistic effects against these bacterial and candida species [59]. In a different study, Oliveira et al., [60] demonstrated that α-bisabolol had antimicrobial activity against S. aureus ATCC 25923, with a minimum inhibitory concentration of 161.27 μg mL−1. Similarly, α-bisabolol in combination with the antibiotic norfloxacin exerted a synergistic antimicrobial effect against S. aureus ATCC 25923, and α-bisabolol had synergistic action against E. coli when combined with gentamicin [60]. L. inermis L. host other compounds that have been reported to show antibacterial activity, including lawsone [53], naphthoquinone derivatives [61] and 3,5′-hydroxyfavone [62].
In this study, we report considerable antibacterial activity of methanol and ethanol extracts of L. inermis L. of different concentrations on isolates of ESBLs-producing strains of Klebsiella pneumoniae, with zone of inhibitions ranging from 7.3 ± 0.6 mm to 13.70 ± 1.2 mm and 6.00 ± 0.6 to 11.00 ± 0.0 mm, respectively. Similar to this study, Moutawalli et al. [63] showed that methanol extract demonstrated a wide range of activity, 11 ± 0.1 mm to 18 ± 0.2 mm, on different bacterial isolates. This could explain the possible broad-spectrum activity of Lawsonia inermis L. Specifically, the results obtained in this study indicate that the methanol extract had the highest activity of 13.7 ± 1.2 mm at 100 mg/mL concentration (Table 3). Furthermore, the high zone of inhibition of the methanol extract was also reported by Nigussie et al. [64]. In a general context, the antibacterial potential of both methanolic and ethanolic extracts of L. inermis L. might be due to the different phytochemicals identified, and this has been confirmed by a previous study [65].
In an attempt to determine the active compounds of L. inermis L., a molecular docking analysis was performed starting with the phytochemicals, physicochemical, pharmacokinetics and docking score. The twenty-seven (27) phytochemicals of the methanol extract of L. inermis L. detected with the aid of GC-MS were evaluated for physicochemical and pharmacokinetic activity in order to determine if the compounds have a drug-likeness. The method employed by Nyalo et. al. [66] and Usman et al. [67] was adopted for evaluating the therapeutic safety, metabolism and accuracy of the compounds identified by GC-MS. Many parameters were used to evaluate the physiochemical properties, including molecular weight, hydrogen-bond acceptor (HBA), hydrogen-bond donor (HBD), logarithms of partial coefficient (LogP) and drug likeness. The interpretation of the physiochemical analysis of the identified compounds was performed according to Lipinski et al. [68] and Egan et al. [69] rules, which is in accordance with the rule of drug-likeness, which is usually employed during the creation of new drugs. The five rules of Lipinski state that for a compound to be termed as a drug, it must have good permeability of the membrane, high gastrointestinal tract absorption, good oral bioavailability and a molecular weight less than or equal to 500 Da (≤500), logarithms of partial coefficient (LogP) less than or equal to 5.88 (≤5.88), a hydrogen-bond donor (HBD) less than or equal to 5 (≤5) and a hydrogen-bond acceptor (HBA) than or equal to 10 (≤10). The Egan rule states that for a compound to possess therapeutic characteristics, it must possess a logarithm of partial coefficient (LogP) less than or equal to 5.88 (≤5.88) and a topological polar surface area (TPSA) of less than or equal to 131 (≤131). Out of the 27 compounds detected, only twelve (12) were revealed to have evidence of drug-likeness activity.
These 12 compounds were then used for molecular docking with a view to examine their free binding energies (docking score) with the Klebsiella pneumoniae CTX-M protein. The molecular docking revealed docking scores ranging from −3.62 kcal/mol to −9.76 kcal/mol. The 12 compounds produced a good docking score because the negative docking score reveals a stronger binding activity between the protein and the ligand. Five of the compounds, CID_5363192, CID_440869, CID_5363411, CID_5283028 and CID_535324, produced residues that include hydrogen bonding. From the result, it can be seen that CID_440869 (2-[1,2-dihydroxyethyl]-9-[β-d-ribofuranosyl] hypoxanthine) has the highest docking score and could be considered as a suitable compound to use for the treatment of infections caused by ESBLs-producing Klebsiella pneumoniae strains.

5. Conclusions

The present study highlights the first combined report on in vitro and in silico studies on the effects of L. inermis L. against multidrug-resistant Klebsiella pneumoniae strains. The results also demonstrate the antibacterial potential of the methanol and ethanol extracts of L. inermis L. on drug-resistant K. pneumoniae. A total of twenty-seven compounds were identified using the GC-MS analysis of the methanol extracts of L. inermis L. The results reveal that of the 27 bioactive compounds identified from the GC-MS analysis, which were screened by evaluating their physicochemical and pharmacokinetic properties, only 12 have drug-likeness properties. The compounds were used for a molecular docking analysis, which revealed the CTX-M protein and CID_440869 to have the highest free binding energy. Therefore, this compound can be considered as a potential therapeutic agent for treating infections caused by strains of ESBLs-producing Klebsiella pneumoniae. Further research could explore the potential antimicrobial applications of L. inermis L. extracts to many bacterial strains.

Author Contributions

A.M., R.K.S. and U.A.E. conceived the study. The study was managed by A.M. and U.A.E. Data curating was performed by A.M., U.A.E., A.N.A., M.A.I. and R.K.S. Data analysis was conducted by A.M., A.N.A., M.A.I. and R.K.S., A.M. and U.A.E. developed the first draft of the document. U.A.E., A.F.A., B.N.U., A.N.A., M.A.I., S.S. and R.K.S. critically reviewed the draft manuscript and made inputs prior to the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data generated or analyzed during this study are included in this published article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mustapha, A.; Allamin, I.A.; Ismail, H.Y.; Eze, U.A.; Ibrahim, M.M.; Bello, R.Y.M.; Faruq, A.U. Environmental contribution to antimicrobial resistance: A largely ignored global health issue. Proc. Niger. Acad. Sci. 2021, 14, 61–86. [Google Scholar] [CrossRef]
  2. De Kraker, M.E.A.; Stewardson, A.J.; Harbarth, S. Will 10 million people die a year due to antimicrobial resistance by 2050? Public Libr. Sci. Med. 2016, 13, e1002184. [Google Scholar] [CrossRef] [PubMed]
  3. Murray, C.J.L.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Aguilar, G.R.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  4. Mustapha, A.; Chidiebere, O.; Victor, F.I.; Bata, M.M.; Tanko, N.; Yakubu, M.N.; Ali, Z.D.; David, A.S. Antibacterial activity of Nigerian medicinal plants as panacea for antibiotic resistance: A systematic review. J. Med. Herbs 2022, 13, 11–23. [Google Scholar]
  5. Asokan, G.V.; Tufoof, R.; Eman, A.; Hala, S. WHO Global Priority Pathogens List: A Bibliometric Analysis of Medline PubMed for Knowledge Mobilization to Infection Prevention and Control Practices in Bahrain. Oman Med. J. 2019, 34, 184–193. [Google Scholar] [CrossRef] [PubMed]
  6. Mustapha, A.; Disa, H.A.; Balaman, N. Antibacterial activity of Salvadora persica (chewing stick) on Streptococcus mutans isolates from patients attending University of Maiduguri Teaching Hospital’s Dental Unit. Indian J. Res. 2017, 4, 715–717. [Google Scholar]
  7. Ishola, A.O.; Mustapha, A.; Eze, U.A. The use of essential oils as anti-infective agents in the treatment of respiratory tract bacterial infections: A systematic review and meta-analysis. Gomal J. Med. Sci. 2023, 21, 50–59. [Google Scholar]
  8. Yang, X.; Li, X.; Qiu, S.; Liu, C.; Chen, S.; Xia, H.; Zeng, Y.; Shi, L.; Chen, J.; Zheng, J.; et al. Global antimicrobial resistance and antibiotic use in COVID-19 patients within health facilities: A systematic review and meta-analysis of aggregated participant data. J. Infect. 2024, 89, 106183. [Google Scholar] [CrossRef]
  9. Langford, B.J.; Soucy, J.R.; Leung, V.; So, M.; Kwan, A.T.H.; Portnoff, J.S.; Bertagnolio, S.; Raybardhan, S.; MacFadden, D.R.; Daneman, N. Antibiotic resistance associated with the COVID-19 pandemic: A systematic review and meta-analysis. Clin. Microbiol. Infect. 2023, 29, 302–309. [Google Scholar] [CrossRef]
  10. Farahmandfar, R.; Esmaeilzadeh, R.R.; Asnaashari, M.; Shahrampour, D.; Bakhshandeh, T. Bioactive compounds, antioxidant and antimicrobial activities of Arum maculatum leaves extracts as affected by various solvents and extraction methods. Food Sci. Nutr. 2019, 7, 465–475. [Google Scholar] [CrossRef]
  11. Wasihun, Y.; Alekaw Habteweld, H.; Dires Ayenew, K. Antibacterial activity and phytochemical components of leaf extract of Calpurnia aurea. Sci. Rep. 2023, 13, 9767. [Google Scholar] [CrossRef]
  12. Mohammed, E.A.A.; Muddathir, A.M.; Osman, M.A. Antimicrobial activity, phytochemical screening of crude extracts, and essential oils constituents of two Pulicaria spp. growing in Sudan. Sci. Rep. 2020, 10, 17148. [Google Scholar] [CrossRef] [PubMed]
  13. Li, J.; Feng, W.; Dai, R.; Li, B. Recent Progress on the Identification of Phenanthrene Derivatives in Traditional Chinese Medicine and Their Biological Activities. Pharmacol. Res.-Mod. Chin. Med. 2022, 3, 100078. [Google Scholar] [CrossRef]
  14. Oladosu, I.A.; Balogun, S.O.; Zhi-Qiang, L. Chemical constituents of Allophylus africanus. Chin. J. Nat. Med. 2015, 13, 133–141. [Google Scholar] [PubMed]
  15. Batiha, G.E.; Teibo, J.O.; Shaheen, H.M.; Babalola, B.A.; Teibo, T.K.A.; Al-kuraish, H.M.; Al-Garbeeb, A.; Alexiou, A.; Papadakis, M. Therapeutic potential of Lawsonia inermis Linn: A comprehensive overview. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 397, 3525–3540. [Google Scholar] [CrossRef]
  16. Fatahi, B.M.; Salary, S.; Mirzaei, F.; Mahmoodian, H.; Meftahizade, H.; Zareshahi, R. Antibacterial and anti-trichomunas characteristics of local landraces of Lawsonia inermis L. BMC Complement. Med. Ther. 2022, 22, 203. [Google Scholar] [CrossRef]
  17. World Health Organization. Legal Status of Traditional Medicine as Complementary and Alternative Medicine. A World Review; WHO: Geneva, Switzerland, 2017. [Google Scholar]
  18. Al-Snaf, A. A review on Lawsonia inermis: A potential medicinal plant. Int. J. Curr. Pharm. Res. 2019, 11, 1–13. [Google Scholar] [CrossRef]
  19. Khazaeli, P.; Mehrabani, M.; Mosadegh, A.; Bios, S.; Moshaf, M.H. Identification of luteolin in henna (Lawsonia Inermis) oil, a Persion medicine product, by HPTLC and evaluating its antimicrobial effects. Res. J. Pharmacogn. 2019, 6, 51–55. [Google Scholar]
  20. Sheikh, B.A. Development of new therapeutics to meet the current challenge of drug-resistant tuberculosis. Curr. Pharm. Biotechnol. 2021, 22, 480–500. [Google Scholar] [CrossRef]
  21. Chang, D.; Sharma, L.; Dela Cruz, C.S.; Zhang, D. Clinical epidemiology, risk factors, and control strategies of Klebsiella pneum infection. Front. Microbiol. 2021, 12, 750662. [Google Scholar] [CrossRef]
  22. Walther-Rasmussen, J.; Høiby, N. Cefotaximases (CTX-M-ases), an expanding family of extended-spectrum β-lactamases. Can. J. Microb. 2004, 50, 137–165. [Google Scholar] [CrossRef] [PubMed]
  23. Celenza, G.; Pellegrini, C.; Caccamo, M.; Segatore, B.; Amicosante, G.; Perilli, M. Spread of bla CTX-M-type and bla PER-2 β-lactamase genes in clinical isolates from Bolivian hospitals. J. Antimicrob. Chemother. 2006, 57, 975–978. [Google Scholar] [CrossRef]
  24. Cantón, R.; González-Alba, J.M.; Galán, J.C. CTX-M enzymes: Origin and diffusion. Front. Microbiol. 2012, 3, 110. [Google Scholar] [CrossRef]
  25. Eze, U.A.; Eze, N.M.; Mustapha, A. Enzymatic inactivation of penicillins: An emerging threat to global public health. Int. J. Pharm. Sci. Res. 2015, 6, 3151–3160. [Google Scholar]
  26. Peirano, G.; Chen, L.; Kreiswirth, B.N.; Pitout, J.D. Emerging antimicrobial-resistant high-risk Klebsiella pneumoniae clones ST307 and ST147. Antimicrob. Agents Chemother. 2020, 64, 10–128. [Google Scholar] [CrossRef]
  27. Saifi, S.; Ashraf, A.; Hasan, G.M.; Shamsi, A.; Hassan, I. Insights into the preventive actions of natural compounds against Klebsiella pneumoniae infections and drug resistance. Fitoterapia 2024, 173, 105811. [Google Scholar] [CrossRef] [PubMed]
  28. Thamer, F.H.; Thamer, N. Gas chromatography—Mass spectrometry (GC-MS) profiling reveals newly described bioactive compounds in Citrullus colocynthis (L.) seeds oil extracts. Heliyon 2023, 9, e16861. [Google Scholar] [CrossRef] [PubMed]
  29. Kumar, D.; Singh, L.; Antil, R.; Kumari, S. GC-MS analysis and phytochemical screening of methanolic fruit extract of Citrullus colocynthis (L.) Schrad. J. Pharmacogn. Phytochem. 2019, 8, 3360–3363. [Google Scholar]
  30. Lee, K.; Kim, D. In-silico molecular binding prediction for human drug targets using deep neural multi-task learning. Genes 2019, 10, 906. [Google Scholar] [CrossRef]
  31. Bharathi, A.; Roopan, S.M.; Vasavi, C.S.; Munusami, P.; Gayathri, G.A.; Gayathri, M. In silico molecular docking and in vitro antidiabetic studies of dihydropyrimido [4, 5-a] acridin-2-amines. BioMed Res. Int. 2014, 2014, 971569. [Google Scholar] [CrossRef]
  32. Seeliger, D.; de Groot, B.L. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J. Comput. Aided Mol. Des. 2010, 24, 417–422. [Google Scholar] [CrossRef] [PubMed]
  33. Isa, M.A.; Mustapha, A.; Qazi, S.; Raza, K.; Allamin, I.A.; Ibrahim, M.M.; Mohammed, M.M. In silico molecular docking and molecular dynamic simulation of potential inhibitors of 3C-like main proteinase (3CLpro) from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) using selected African medicinal plants. Adv. Tradit. Med. 2022, 22, 107–123. [Google Scholar] [CrossRef]
  34. Zannat, K.E.; Saha, S.K.; Tanzim, S.M.; Afrin, A.; Saha, B.C.; Joynal, J.B.; Aktar, M.; Nira, N.H.; Akhter, N.; Hossain, M.A. Antibacterial Effects of Chloroform Henna (Lawsonia inermis) Leaf Extracts against Two Nosocomial Infection Causing Pathogens: Gram-positive Staphylococcus aureus and Gram-negative Klebsiella pneumoniae: A Comparative Study. Mymensingh Med. J. MMJ 2023, 32, 620–626. [Google Scholar]
  35. Arun, P.; Purushotham, K.G.; Jayarani, J.; Kumara, V. In vitro Antibacterial activity and Flavonoid contents of Lawsonia inermis (Henna). Int. J. PharmTech Res. 2010, 2, 1178–1181. [Google Scholar]
  36. Pasandi, P.A.; Farahbakhsh, H. Lawsonia inermis L. leaves aqueous extract as a natural antioxidant and antibacterial product. Nat. Prod. Res. 2020, 34, 3399–3403. [Google Scholar] [CrossRef]
  37. Abdallah, E.M.; Aldamegh, M.A. Antibacterial and Toxicological assessment of Lawsonia inermis Linn. (Henna) leaves on Rat. Res. J. Biol. Sci. 2011, 6, 275–280. [Google Scholar]
  38. El-Kamali, H.H.; EL-Karim, E.M.W. Evaluation of Antibacterial Activity of Some Medicinal Plants Used in Sudanese Traditional Medicine for Treatment of Wound Infections. Acad. J. Plant Sci. 2009, 2, 246–251. [Google Scholar]
  39. Idris, A.H.; Haruna, Z.; Iliyasu, M.Y.; Sahal, M.R.; Inusa, T.; Salisu, A.; Isma’il, S.; Umar, R.D.; Kabeer, Z.M.; Tahir, H.; et al. Antibacterial Activity of Lawsonia inermis Leaf Extracts against Multidrug-resistant Pseudomonas aeruginosa from Infected Wounds. Eur. J. Med. Plants 2023, 34, 1–8. [Google Scholar] [CrossRef]
  40. Cheesbrough, M. (Ed.) Biochemical tests to identify bacteria. In Laboratory Practice in Tropical Countries; Cambridge Edition; Cambridge University Press: Cambridge, UK, 2002; pp. 36–70. [Google Scholar]
  41. Nabi, M.; Zargar, M.I.; Tabassum, N.; Ganai, B.A.; Wani, S.U.D.; Alshehri, S.; Alam, P.; Shakeel, F. Phytochemical profiling and antibacterial activity of methanol leaf extract of Skimmia anquetilia. Plants 2022, 11, 1667. [Google Scholar] [CrossRef]
  42. Gul, R.; Jan, S.U.; Faridullah, S.; Sherani, S.; Jahan, N. Preliminary phytochemical screening, quantitative analysis of alkaloids, and antioxidant activity of crude plant extracts from Ephedra intermedia indigenous to Balochistan. Sci. World J. 2017, 2017, 5873648. [Google Scholar] [CrossRef]
  43. Wakil, S.; Isa, M.A.; Mustapha, A. Phenotypic and Molecular Detection of Resistance Genes from Multi-drug-Resistant Escherichia coli Isolated from Patients Attending Selected Hospitals in Damaturu, Yobe State, Nigeria. Int. J. Res. Rev. 2021, 8, 396–407. [Google Scholar] [CrossRef]
  44. CLSI. Performance Standards for Antimicrobial Susceptibility Testing, 26th ed.; M.1.0.0.S.; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2016. [Google Scholar]
  45. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef]
  46. Johansson, M.U.; Zoete, V.; Michielin, O.; Guex, N. Defining and searching for structural motifs using DeepView/Swiss-PdbViewer. BMC Bioinform. 2012, 13, 173. [Google Scholar] [CrossRef]
  47. Ramachandran, S.; Kota, P.; Ding, F.; Dokholyan, N.V. Automated minimization of steric clashes in protein structures. Proteins Struct. Funct. Bioinform. 2011, 79, 261–270. [Google Scholar] [CrossRef]
  48. Sander, T.; Freyss, J.; von Korff, M.; Rufener, C. Datawarrior: An open-source program for Chemistry aware data visualization and analysis. J. Chem. Inf. Model. 2015, 55, 460–473. [Google Scholar] [CrossRef] [PubMed]
  49. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef]
  50. Cheng, F.; Li, W.; Zhou, Y.; Shen, J.; Wu, Z.; Liu, G.; Lee, P.W.; Tang, Y. AdmetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. J. Chem. Inf. Model. 2012, 52, 3099–3105. [Google Scholar] [CrossRef]
  51. Morris, G.M.; Huey, R.; Lindstorm, 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. 1998, 30, 2785–2791. [Google Scholar] [CrossRef] [PubMed]
  52. Hariono, M.; Abdullah, N.; Damodaran, K.V.; Kamarulzaman, E.E.; Mohamed, N.; Hassan, S.S.; Shamsuddin, S.; Wahab, H.A. Potential new H1N1 neuraminidase inhibitors from ferulic acid and vanillin: Molecular modelling, synthesis and in vitro assay. Sci. Rep. 2016, 6, 38692. [Google Scholar] [CrossRef]
  53. Jeyaseelan, E.C.; Jenothiny, S.; Pathmanathan, M.K.; Jeyadevan, J.P. Antibacterial activity of sequentially extracted organic solvent extracts of fruits, flowers and leaves of Lawsonia inermis L. from Jaffna. Asian Pac. J. Trop. Biomed. 2012, 2, 798–802. [Google Scholar] [CrossRef]
  54. Popova, V.; Ivanova, T.; Stoyanova, A.; Nikolova, V.; Hristeva, T.; Gochev, V.; Yonchev, Y.; Nikolov, N.; Zheljazkov, V.D. Terpenoids in the essential oil and concentrated aromatic products obtained from Nicotiana glutinosa L. Leaves. Molecules 2019, 25, 30. [Google Scholar] [CrossRef] [PubMed]
  55. Augostine, C.R.; Avery, S.V. Discovery of natural products with antifungal potential through combinatorial synergy. Front. Microbiol. 2022, 13, 866840. [Google Scholar] [CrossRef] [PubMed]
  56. Iobbi, V.; Brun, P.; Bernabé, G.; Dougue Kentsop, R.A.; Donadio, G.; Ruffoni, B.; Fossa, P.; Bisio, A.; De Tommasi, N. Labdane diterpenoids from Salvia tingitana Etl. synergize with clindamycin against methicillin-resistant Staphylococcus aureus. Molecules 2021, 26, 6681. [Google Scholar] [CrossRef]
  57. Chen, Q.; Tang, K.; Guo, Y. Discovery of sclareol and sclareolide as filovirus entry inhibitors. J. Asian Nat. Prod. Res. 2020, 22, 464–473. [Google Scholar] [CrossRef]
  58. Eddin, L.B.; Jha, N.K.; Goyal, S.N.; Agrawal, Y.O.; Subramanya, S.B.; Bastaki, S.M.; Ojha, S. Health benefits, pharmacological effects, molecular mechanisms, and therapeutic potential of α-bisabolol. Nutrients 2022, 14, 1370. [Google Scholar] [CrossRef] [PubMed]
  59. Rodrigues, F.F.G.; Colares, A.V.; Nonato, C.F.A.; Galvão-Rodrigues, F.F.; Mota, M.L.; Moraes Braga, M.F.B.; Costa, J. In vitro antimicrobial activity of the essential oil from Vanillosmopsis arborea Barker (Asteraceae) and its major constituent, α-Bisabolol. Microb. Pathog. 2018, 125, 144–149. [Google Scholar] [CrossRef]
  60. Oliveira, F.S.; de Freitas, T.S.; da Cruz, R.P.; do Socorro Costa, M.; Pereira, R.L.S.; Quintans-Júnior, L.J.; de Araújo Andrade, T.; dos Passos Menezes, P.; de Sousa, B.M.H.; Nunes, P.S.; et al. Evaluation of the antibacterial and modulatory potential of α-Bisabolol, β-cyclodextrin and α-Bisabolol/β-cyclodextrin complex. Biomed. Pharm. 2017, 92, 1111–1118. [Google Scholar] [CrossRef]
  61. Abbas, Z.; Siddiqui, B.S.; Shahzad, S.; Sattar, S.; Begum, S.; Batool, A.; Choudhary, M.I. Lawsozaheer, a new chromone produced by an endophytic fungus Paecilomyces variotii isolated from Lawsonia Alba Lam. inhibits the growth of Staphylococcus aureus. Nat. Prod. Res. 2020, 35, 4448–4453. [Google Scholar] [CrossRef]
  62. Yang, C.-S.; Chen, J.-J.; Huang, H.-C.; Huang, G.-J.; Wang, S.-Y.; Sung, P.-J.; Cheng, M.-J.; Wu, M.-D.; Kuo, Y.-H. New benzenoid derivatives and other constituents from Lawsonia inermis with inhibitory activity against NO production. Molecules 2017, 22, 936–944. [Google Scholar] [CrossRef]
  63. Moutawalli, A.; Benkhouili, F.Z.; Doukkali, A.; Benzeid, H.; Zahidi, A. The biological and pharmacologic actions of Lawsonia inermis L. Phytomed. Plus 2023, 3, 100468. [Google Scholar] [CrossRef]
  64. Nigussie, D.; Davey, G.; Legesse, B.A.; Fekadu, A.; Makonnen, E. Antibacterial activity of methanol extracts of the leaves of three medicinal plants against selected bacteria isolated from wounds of lymphoedema patients. BMC Complement. Med. Ther. 2021, 21, 2. [Google Scholar] [CrossRef]
  65. Nimbeshaho, F.; Mwangi, C.; Orina, F.; Chacha, M.; Adipo, N.; Moody, J.; Kigondu, E. Antimycobacterial activities, cytotoxicity and phytochemical screening of extracts for three medicinal plants growing in Kenya. J. Med. Plants Res. 2020, 14, 129–143. [Google Scholar]
  66. Nyalo, P.O.; Omwenga, G.I.; Ngugi, M.P. Antibacterial activity and GC-MS analysis of ethyl acetate extracts of Xerophyta spekei (Baker) and Grewia tembensis (Fresen). Heliyon 2023, 9, e14461. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Usman, R.A.; Rabiu, U. Antimicrobial activity of Lawsonia inermis (henna) extracts. Bayero J. Pure Appl. Sci. 2006, 11, 167–671. [Google Scholar] [CrossRef]
  68. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeny, P. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  69. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of drug absorption using multivariate statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef]
Figure 1. Chromatogram of the gas chromatography–mass spectroscopy analysis performed on the methanolic extract of the Lawsonia inermis leaves (GC 7890B, MSD 5977A, Agilent Tech, Santa Clara, CA, USA).
Figure 1. Chromatogram of the gas chromatography–mass spectroscopy analysis performed on the methanolic extract of the Lawsonia inermis leaves (GC 7890B, MSD 5977A, Agilent Tech, Santa Clara, CA, USA).
Biomed 04 00022 g001
Table 1. Phytochemicals of Lawsonia inermis L.
Table 1. Phytochemicals of Lawsonia inermis L.
PhytochemicalMethanolEthanol
Alkaloids++
Tannins++
Flavonoids++
Anthraquinones++
+ indicates positive result for the phytochemical.
Table 2. Compounds obtained from the GC–MS analysis of Lawsonia inermis L.
Table 2. Compounds obtained from the GC–MS analysis of Lawsonia inermis L.
S/NPubChem IDCompoundFormulaMolecular WeightRetention Time (min)Peaks
16590Ethyl acetateC4H8O2883.7161
2536425Heptane, 4-azidoC7H15N31414.8172
3225038Pentyl glycolateC7H14O31464.8172
476029Propanoic acid, 2-(aminooxy)-C3H7NO31055.6893
553631927-Hydroxy-3-(1,1-dimethylprop-2-enyl) coumarinC14H14O32305.6893
6543621Pentanoic acid, 2-(aminooxy)-C5H11NO31335.6893
74408692-[1,2-Dihydroxyethyl]-9-[β-d-ribofuranosyl] hypoxanthine C12H16N4O73285.6893
85369802-Heptanone, 6-methyl-5-methylene-C9H16O1406.0484
95345924,5,9-Trihydroxy-dodeca-1,11-dieneC12H22O32146.6755
1053631927-Hydroxy-3-(1,1-dimethylprop-2-enyl) coumarinC14H14O32307.0676
115364118,8-Dimethyl-7,9-dioxabicyclo [4.3.0] nonane-3-carboxylicacid, methyl esterC11H18O42147.0676
125283028trans-Traumatic acidC12H20O42287.0676
135372889,9-Dimethoxybicyclo [3.3.1] nona-2,4-dioneC11H16O42127.2997
148180Undecanoic acidC11H22O21867.2997
1511996452ViridiflorolC15H26O2227.9668
168842CitronellolC10H20O1568.0699
1764212992,2,3,3,4,4-HexamethyltetrahydrofuranC10H20O1568.39511
181632631-Naphthalenepropanol, α-ethenyldecahydro-5-(hydroxymethyl)-α,2,5,5,8a-pentamethyl-C20H36O230810.60613
195367736Cyclopropanol, 1-(3,7-dimethyl-1-octenyl)-C13H24O19612.23515
20535324Bicyclo [3.2.1]oct-3-en-2-one, 3,8-dihydroxy-1-methoxy-7-(7-methoxy-1,3-benzodioxol-5-yl)-6-methyl-5-(2-propenyl)-, [1R-(6-endo,7-exo,8-syn)]-C21H24O738812.23515
2153632741,2-dihydro-8-hydroxylinalool C10H20O217213.91616
22101282029Bicyclo [3.2.1]oct-3-en-2-one, 3,8-dihydroxy-1-methoxy-7-(7-methoxy-1,3-benzodioxol-5-yl)-6-methyl-5-(2-propenyl)-, [1R-(6-endo,7-exo,8-syn)]-C21H24O722518.17418
2353658314,4,8-Trimethyl-non-5-enalC12H22O18218.17418
243190687713-Cyclohexene-1-methanol, α,4-dimethyl-α-(4-methyl-3-pentenyl)-, [R-(R*,R*)]-C15H26O22218.17418
25296248photocitral BC10H16O15220.43619
265513001,2-Pentanediol, 5-(6-bromodecahydro-2-hydroxy-2,5,5a,8a-tetramethyl-1-naphthalenyl)-3-methylene-C20H35BrO340222.91520
275371182-Isopropyl-5-methylcyclohexyl 3-(1-(4-chlorophenyl)-3-oxobutyl)-coumarin-4-yl carbonateC30H33ClO652524.46522
Table 3. Zone of inhibition (mm) of methanol and ethanol extract of Lawsonia inermis L. extract against multidrug-resistant Klebsiella pneumoniae.
Table 3. Zone of inhibition (mm) of methanol and ethanol extract of Lawsonia inermis L. extract against multidrug-resistant Klebsiella pneumoniae.
Extract/Zone of Inhibition (mm)
Concentration (mg/mL)EthanolMethanol
2511.0 ± 0.0 bc11.3 ± 0.6 bc
506.3 ± 0.6 e12.0 ± 0.0 b
758.0 ± 1.0 d7.3 ± 0.6 de
10010.0 ± 1.0 c13.7 ± 1.2 a
Values indicate mean ± standard deviation (SD). Values with the same letter(s) are not significantly different using Tukey multiple range test at p < 0.05.
Table 4. Physiochemical analysis of compounds from Lawsonia inermis L. identified using GC-MS.
Table 4. Physiochemical analysis of compounds from Lawsonia inermis L. identified using GC-MS.
S/NPubChem IDMolecular
Weight (≤500)
Number of
HBA (≤10)
Number of
HBD (≤5)
MolLogP (≤5)Drug Likeness
1CID_659088.1055210.3736−2.82
2CID_536425388.415722.6761−0.89897
3CID_225038146.185310.9716−10.318
4CID_76029105.09342−1.3119−1.4365
5CID_5363192230.262313.0993−4.5992
6CID_543621133.14642−0.4031−3.384
7CID_440869328.280116−3.38267.1752
8CID_536980140.225102.8354−11.248
9CID_534592214.304332.0848−7.1915
10CID_5363192230.262313.0993−4.5992
11CID_536411214.260400.9337−4.8099
12CID_5283028228.287422.7164−15.097
13CID_537288212.244400.8154−16.496
14CID_8180186.294213.7905−25.216
15CID_11996452222.370113.2008−1.9678
16CID_8842156.268113.3494−8.6831
17CID_6421299156.268102.3703−13.972
18CID_163263308.504224.6198−8.3601
19CID_5367736196.333113.8371−2.2587
20CID_535324388.415722.6761−0.89897
21CID_5363274172.267222.4899−1.7531
22CID_101282029388.415722.6761−0.89897
23CID_5365831182.306103.7692−6.1572
24CID_319068771222.370114.4711−1.4665
25CID_296248152.236101.676−7.016
26CID_551300403.399334.3349−11.802
27CID_537118525.039607.3414−22.803
Table 5. Pharmacokinetic analysis of the phyto-compounds detected from Lawsonia inermis L.
Table 5. Pharmacokinetic analysis of the phyto-compounds detected from Lawsonia inermis L.
S/NCompound NameBBBCYP2D6 InhibitorHIAMutagensTumorigenesisReproductive ToxicityIrritant
1CID_6590---NoneNoneNoneHigh
2CID_536425---NoneNoneNoneNone
3CID_225038---LowNoneNoneLow
4CID_76029---HighNoneNoneNone
5CID_5363192---NoneNoneNoneNone
6CID_543621---NoneNoneNoneNone
7CID_440869---NoneNoneNoneNone
8CID_536980---NoneNoneNoneHigh
9CID_534592---NoneNoneNoneLow
10CID_5363192---NoneNoneNoneNone
11CID_536411---NoneNoneNoneNone
12CID_5283028---NoneNoneNoneNone
13CID_537288---NoneNoneNoneNone
14CID_8180---NoneNoneNoneHigh
15CID_11996452---NoneHighNoneHigh
16CID_8842---NoneNoneHighHigh
17CID_6421299---NoneNoneNoneNone
18CID_163263---NoneNoneNoneLow
19CID_5367736---NoneNoneNoneNone
20CID_535324---NoneNoneNoneNone
21CID_5363274---NoneNoneNoneHigh
22CID_101282029---NoneNoneNoneNone
23CID_5365831---HighNoneNoneHigh
24CID_319068771---NoneNoneNoneHigh
25CID_296248---NoneNoneNoneHigh
26CID_551300---HighNoneNoneLow
27CID_537118---NoneNoneHighHigh
Table 6. Docking scores and residues involved in H-bond formation.
Table 6. Docking scores and residues involved in H-bond formation.
S/NoCompound IDDocking Score (kcal/mol)Residues Involve in H-BondsDistance (Ǻ)
1CID_536425−4.98
2CID_5363192−6.00Leu200, Pro201, Gln197
Val206
2.79, 2.70, 2.78, 2.67
2.94
3CID_543621−3.62
4CID_440869−9.76Gly262 Lys2573.09, 2.89
5CID_5363192−6.00Leu200, Gln197, Pro201, Val2062.83, 2.76, 2.75, 2.67
6CID_536411−5.85Gly2622.90
7CID_5283028−5.74Lys227, Arg229, Trp204, Gly199
Lys257
2.68, 3.22, 2.62, 3.18
2.71, 2.75
8CID_537288−4.62
9CID_6421299−4.54
10CID_5367736−4.96
11CID_535324−6.11Trp204
Val206
2.84
2.77
12CID_101282029−4.98
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mustapha, A.; AlSharksi, A.N.; Eze, U.A.; Samaila, R.K.; Ukwah, B.N.; Anyiam, A.F.; Samarasinghe, S.; Ibrahim, M.A. Phytochemical Composition, In Silico Molecular Docking Analysis and Antibacterial Activity of Lawsonia inermis Linn Leaves Extracts against Extended Spectrum Beta-Lactamases-Producing Strains of Klebsiella pneumoniae. BioMed 2024, 4, 277-292. https://doi.org/10.3390/biomed4030022

AMA Style

Mustapha A, AlSharksi AN, Eze UA, Samaila RK, Ukwah BN, Anyiam AF, Samarasinghe S, Ibrahim MA. Phytochemical Composition, In Silico Molecular Docking Analysis and Antibacterial Activity of Lawsonia inermis Linn Leaves Extracts against Extended Spectrum Beta-Lactamases-Producing Strains of Klebsiella pneumoniae. BioMed. 2024; 4(3):277-292. https://doi.org/10.3390/biomed4030022

Chicago/Turabian Style

Mustapha, Adam, Ahmed Nouri AlSharksi, Ukpai A. Eze, Rahma Kudla Samaila, Boniface Nwofoke Ukwah, Arinze Favour Anyiam, Shivanthi Samarasinghe, and Musa Adamu Ibrahim. 2024. "Phytochemical Composition, In Silico Molecular Docking Analysis and Antibacterial Activity of Lawsonia inermis Linn Leaves Extracts against Extended Spectrum Beta-Lactamases-Producing Strains of Klebsiella pneumoniae" BioMed 4, no. 3: 277-292. https://doi.org/10.3390/biomed4030022

Article Metrics

Back to TopTop