You are on page 1of 6

Food and Chemical Toxicology 154 (2021) 112348

Contents lists available at ScienceDirect

Food and Chemical Toxicology


journal homepage: www.elsevier.com/locate/foodchemtox

A comprehensive phytochemical, biological, toxicological and molecular


docking evaluation of Suaeda fruticosa (L.) Forssk.: An edible halophyte
medicinal plant
Hammad Saleem a, b, *, Umair Khurshid c, Muhammad Sarfraz d, Muhammad Imran Tousif e,
Abdulwahab Alamri f, Sirajudheen Anwar f, Abdulhakeem Alamri g, Irshad Ahmad h,
Hassan H. Abdallah i, Fawzi M. Mahomoodally j, Nafees Ahemad b
a
Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
b
School of Pharmacy, Monash University, Jalan Lagoon Selatan, 47500 Bandar Sunway Selangor Darul Ehsan, Malaysia
c
Bahawalpur College of Pharmacy, Bahawalpur Medical and Dental College, Bahawalpur, Pakistan
d
College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
e
Department of Chemistry, Township Campus, University of Education, Lahore, Pakistan
f
Department of Pharmacology & Toxicology, College of Pharmacy, University of Hail, Saudi Arabia
g
Department of Clinical Laboratory Sciences, College of Applied Medical Science, Taif University, P. O. Box 11099, Taif, 21944, Saudi Arabia
h
Department of Pharmacy, The Islamia University of Bahawalpur, Pakistan
i
Chemistry Department, College of Education, Salahaddin University, Erbil, Iraq
j
Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Mauritius

A R T I C L E I N F O A B S T R A C T

Handling Editor: Dr. Jose Luis Domingo Suaeda fruticosa is an edible medicinal halophyte known for its traditional uses. In this study, methanol and
dichloromethane extracts of S. fruticosa were explored for phytochemical, biological and toxicological parame­
Keywords: ters. Total phenolic and flavonoid constituents were determined by using standard aluminum chloride and Folin-
Suaeda fruticose Ciocalteu methods, and UHPLC-MS analysis of methanol extract was performed for tentative identification of
Phytochemicals
secondary metabolites. Different standard methods like DPPH, ABTS, FRAP, CUPRAC, total antioxidant capacity
Antioxidant
(TAC), and metal chelation assays were utilized to find out the antioxidant potential of extracts. Enzyme inhi­
Enzyme inhibition
Cytotoxicity bition studies of extracts against acetylcholinesterase, butyrylcholinesterase, tyrosinase, α-amylase and,
Docking α-glucosidase enzymes were also studied. Likewise, the cytotoxicity was also assessed against MCF-7, MDA-MB-
231, and DU-145 cell lines. The higher phenolic and flavonoids contents were observed in methanol extracts
which can be correlated to its higher radical scavenging potential. Similarly, 11 different secondary metabolites
were tentatively identified by UHPLC profiling. Both the extract showed significant inhibition against all the
enzymes except for α-glucosidase. Moreover, docking studies were also performed against the tested enzymes. In
the case of cytotoxicity, both the samples were found moderately toxic against the tested cell lines. This plant can
be explored further for its potential therapeutic and edible uses.

1. Introduction For example, an edible halophyte plant named Mesembryanthemum edule


is a traditional remedy for bacterial and fungal infections and also used
Halophytes have the greater ability to survive and bear toxic reactive as a treatment for diarrhoea, sinusitis, infantile eczema, and tuberculosis
oxygen species (ROS) due to a very strong antioxidant systems, (van der Watt and Pretorius, 2001), likewise Tamarix gallica is utilized as
including enzymatic and non-enzymatic components (Ksouri et al., a detergent, diuretic, expectorant, laxative, and astringent (Ksouri et al.,
2009). The literature data indicates that a number of medicinal halo­ 2009).
phytes have been reported to possess important bioactive compounds, S. Fruticosa is also known as shrubby seablite, belongs to the genus
including polyphenols which are helpful for different remedial purposes. Suaeda of the family Amaranthaceae. This plant is widely distributed

* Corresponding author. Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan.
E-mail address: hammad.saleem@uvas.edu.pk (H. Saleem).

https://doi.org/10.1016/j.fct.2021.112348
Received 23 May 2021; Received in revised form 7 June 2021; Accepted 12 June 2021
Available online 16 June 2021
0278-6915/© 2021 Elsevier Ltd. All rights reserved.
H. Saleem et al. Food and Chemical Toxicology 154 (2021) 112348

over the coasts of northern Africa, the Mediterranean region, Atlantic 2.2. Phytochemical composition
coasts of southern Spain, Portugal, France, south-eastern England, Iran,
Afghanistan, and the Indian sub-continent (Jalas and Suominen, 1988). 2.2.1. Total phenolic and flavonoid contents
S. fruticosa is a medicinal and edible halophyte that can be grown under Total phenolic (TPC) and total flavonoid (TFC) contents were
saline conditions (Ksouri et al., 2009). It commonly grows in flooded determined via previously described standard Folin-Ciocalteu and
alluvial areas, drier areas, salt marshes, salt flats, and coastal areas. It is aluminum chloride protocols, respectively (Yadav and Agarwala, 2011;
also found over the saline regions in southern Africa (Goodall et al., Zengin et al., 2016). The results of TPC were noted as mg GAE/g of
2009). The seeds and young shoots of this plant have been reported to be extract (milligrams of gallic acid equivalent), while the findings of TFC
consumed (cooked or raw) by humans (Wickens et al., 2012). The seeds were recorded as mg QE/g extract (milligrams of quercetin equivalent).
contain a high-quality edible oil that is abundant in unsaturated fatty
acids (Weber et al., 2007). The plant can be utilized to reduce soil 2.2.2. UHPLC-MS analysis
salinity and to remediate hazardous metal-contaminated soils (Hameed Secondary metabolites of methanol extract were determined UHPLC-
et al., 2012). The plants’ high salt content will most likely limit their MS analysis as describe previously (Saleem et al., 2019). The details of
usage as stand-alone fodder crops; instead, they will most likely be UHPLC-MS instrumentation are provided in the supplementary material
employed as components of a feed mix (Ahmad and Malik, 2013). section.
The different parts of this plant have hypolipidaemic, hypoglycemic,
and antiophthalmic effects (Bennani-Kabchi et al., 1999). S. fruticosa
cultivation may help in bioremediation and reclamation of the soil that 2.3. Biological activities
is contaminated with salinity and toxic metal (Bareen and Tahira, 2011).
S fruticosa contains a large number of compounds like chromo alkaloids 2.3.1. Antioxidant activities
which contain betacyanins and betaxanthine, collectively known as Antioxidant properties of S. fruticosa extracts were estimated by
betalains (Naija et al., 2014). This plant also contains tannins, saponins, using previously described standard protocols, including free radical
terpenoids, coumarins, fatty acids like palmitic acid, oleic acid, linolenic scavenging (DPPH and ABTS), reducing antioxidant power (FRAP and
acid, and polyphenols (Ashraf et al., 2016). S. fruticosa can be used for CUPRAC), total antioxidant capacity (phosphomolybdenum), and metal
wound healing. Its leaves are used to treat ophthalmia. This plant also chelating power assays (Grochowski et al., 2017). The results of all the
acts as a diuretic and laxative. It is also used for various other disorders antioxidant assays were reported as mg TE/g extract, except for the
such as pain, fever, skin, respiratory, toothache, digestive, and genitor metal chelation assay for which the results were presented as mg
urinary disorders (Qasim et al., 2011). EDTAE/g extract. The detailed methodology of antioxidant assays is
Taking into account the medicinal importance of this halophyte provided in the supplementary material section.
edible plant, the present work was designed to evaluate the detailed
phytochemical, biological, and toxicological potential of methanol of 2.3.2. Enzyme inhibition studies
dichloromethane (DCM) extracts of S. fruticosa whole plant. The The studied plant extracts were tested against different enzymes,
phytochemical composition was ascertained by determining total including acetylcholinesterase (AChE), butyrylcholinesterase (BChE),
phenolic and total flavonoid contents, and ultra-high performance liquid tyrosinase, α-glucosidase, and α-amylase by using in vitro standard
chromatography mass-spectrometry (UHPLC-MS) secondary metabolite methods as detailed earlier (Abirami et al., 2014; Mollica et al., 2017).
analysis. For the biological evaluation, antioxidant and enzyme inhibi­ The standard used for AChE and BChE was galantamine, and their
tion assays were performed. Antioxidant potential was evaluated via inhibitory activity was measured as mg GALAE/g extract (milligrams of
free radical scavenging (2,2-diphenyl-1-picrylhydrazyl -DPPH and 2,2′ - galantamine equivalent per gram of extract). Likewise, acarbose was
azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) -ABTS), reducing used as a standard for α-glucosidase and α-amylase inhibition, and the
power (ferric reducing antioxidant power -FRAP and cupric reducing results were expressed in millimoles of acarbose equivalent per gram of
antioxidant capacity -CUPRAC), total antioxidant power (phosphomo­ extract (mmol ACAE/g extract). For tyrosinase inhibition, kojic acid was
lybdenum), and metal chelation assays. Similarly, the enzyme inhibition employed as standard, and results were recorded as milligrams of kojic
studies against the clinically important enzymes involved in the com­ acid equivalents per gram of extract (mg KAE/g extract). The detailed
mon pathologies, including neurological disorders (acetylcholinesterase methodology of enzyme assays is provided in the supplementary mate­
-AChE and butyrylcholinesterase -BChE), diabetes (α-amylase and rial section.
α-glucosidase), and skin problems (tyrosinase) were performed. The
cytotoxicity of both the extracts was done using MTT assay against the 2.3.3. Cytotoxicity assay
MCF-7, MDA-MB-231 breast cancer cell lines, and DU-145 prostate cell The cytotoxicity activity of the tested samples was tested against two
line. In addition, in order to highlight the interaction and mechanism of breast cancer cell lines, i.e., MDA-MB 231 and MCF-7 cells employing
enzyme inhibition, the major phytochemicals as tentatively identified in the previously described method, with slight modifications (Nem­
the methanol extract were further studies for in-silico molecular docking. udzivhadi and Masoko, 2014). The cell viability percentage (%) after 72
h at the concentration of 200 μg/mL was determined as follows:
2. Material and methods
Percentage cell viability = ABSs – ABSc × 100

2.1. Plant collection and extraction Where ABSs: absorbance of the sample; ABSc: absorbance of control.

The whole plant of S. fruticosa was collected from the peripheral 2.3.4. In-Silico studies/Docking calculations
areas of Bahawalpur, Pakistan. The plant collected was dried under Three dominant compounds were selected for docking studies,
shade for few days and grounded to powder form. The dried powdered namely, 3,4-dihydroxybenzoic acid, 3-O-acetylhamayne, and uplandi­
plant material was extracted using the maceration technique with DCM cine. The starting structures of the three compounds were found online
and methanol solvents (for 72 h with each solvent). A rotary evaporator from the zinc database (Irwin et al., 2012). AM1 semi-emperical
was used to concentrate these resulting extracts. method, which is implemented in gaussian09 software, was used to
optimize the 3D structure to the ground state energy. The optimized
structures were docked at the active site of five enzymes, namely, AChE,
BChE, tyrosinase, α-amylase, and α-glucosidase enzymes. The co­
ordinates of these enzymes were extracted from the crystal structures of

2
H. Saleem et al. Food and Chemical Toxicology 154 (2021) 112348

these enzymes in Protein Databank RCSB PDB. PDB structure with the 3.2. UHPLC-MS phytochemical analysis
code: 4EY6 was used as the AChE receptor in which the enzyme was
crystallized in complex with galantamine. PDB code:1P0P is represent­ Ultra-high-pressure liquid chromatography (UHPLC) is a system
ing the crystal structure of the BChE in complex with the inhibitor working at ultra-high pressure and has decreased the time of analysis for
butyrylthiocholine at the active site of the enzyme. Similarly, the PDB different compounds. This technique is widely used for profiling sec­
code: 5I38 was used as the crystal structure of the tyrosinase enzyme as a ondary metabolites (Grata et al., 2009). In the present study, for the
complex with kojic acid. PDB codes 7TAA and 3W37 were used to tentative identification of different phytocompounds in methanol
represent the enzymes α-amylase and α-glucosidase, respectively. Both extract of S. fruticose, negative ionization mode of UHPLC-MS analysis
enzymes were crystallized as a complex with the acarbose inhibitor at was employed. The total ion chromatogram (TIC) with different peaks of
the active site. Prior to docking, inhibitors, co-crystallized molecules, the tentatively identified secondary metabolites as in methanol extract is
and all water molecules were removed from the retrieved structures. represented in Fig. 1. A total of 11 different secondary metabolites
Autodock4 software (Molinspiration Database) was used to add polar belonging to different classes of phytochemicals were tentatively iden­
hydrogens, neutralize the protein structures using Kollman united atom tified, and the list of these compounds is given in Table 2. Two phenolic
charges and to perform the docking calculations. Docking the three in­ compounds were found, namely 3, 4-dihydrobenzoic acid, and gingerol.
hibitors was performed in a 60 × 60 x 60 grid box with 0.375 Å distance Three compounds, namely pitheduloside B, 5, 8, 12-trihydroxy-9-
between points using Lamarckian genetic algorithm (LGA) for the pre­ octadecenoic acid, and kamahine C were also tentatively identified as
diction of binding free energies. Two hundred fifty conformations were saponin, fatty acid, and spiroketal, respectively. Similarly, D-threonic
produced for each enzyme-inhibitor complex and the control ligand. The acid (carbohydrate) and allantoin (urea derivative) were also identified.
docked conformations were ranked into clusters based on the binding Likewise, two alkaloidal compounds, i.e., uplandicine and 3-O-acetylha­
free energy (ΔG). Discovery studio 5.0 visualizer was used to visualize to mayne were also tentatively identified.
docked inhibitors at the active site and identify the intermolecular in­
teractions with the active site. 3.3. Antioxidant activities

2.4. Statistical analysis Reactive oxygen species (ROS) are radicals with unpaired electrons
that can damage other biomolecules. They are also known as pro-
All experiments were done in triplicates, and their mean values were oxidants. ROS are responsible for causing damage to biomolecules
calculated. The results obtained were expressed as mean values with including as proteins, lipids, carbohydrates, fatty acids, and nucleic
their standard deviation (SD). For the analysis of data, SPSS software acids. They are also capable of causing DNA damage, which is the major
was used. One-way analysis of variance (ANOVA) and Tukey’s test were cause of mutations (Halliwell, 1990) and may also cause different
employed to analyze the results. A statistically significant value of p < common diseases (Alho and Leinonen, 1999; Duh, 1998). Antioxidants
0.05 was obtained. are becoming increasingly important because they block the oxidation
of other molecules, hence preventing the formation of free radicals
(Halliwell, 1995).
3. Results and discussion
In the present work, the antioxidant potential of methanol and DCM
extracts S. fruticose were determined by using different six different
3.1. Total phenolic and flavonoid contents
antioxidant assays, including free radical scavenging (DPPH and ABTS),
reducing power (FRAP and CUPRAC), total antioxidant capacity
Plants are responsible for the production of a large number of organic
(phosphomolybdenum), and metal chelation assays and the results are
compounds called primary and secondary metabolites (Zengin et al.,
assembled in Table 3. The higher radical scavenging antioxidant values
2019). Both primary and secondary metabolites are of great importance,
were observed in the case of methanol extract (DPPH: 9.06 mg TE/g
as the primary metabolites are directly involved in respiration, photo­
extract; ABTS:16.46 mg TE/g extract). Similarly, the methanol extract
synthesis, development, and growth. In contrast, the secondary metab­
was also found most active for the CUPRAC assay, while the DCM extract
olites play an important role in defensive mechanism against herbivores
showed the highest FRAP value. Likewise, the phosphomolybdenum
in plants, provide protection against UV light, also used as signal mol­
assay was carried out to find out the total antioxidant capacity, and the
ecules, dyes, fibers, glues, oils, waxes, drugs, perfumes, flavouring
higher value of antioxidant capacity was exhibited by the DCM extract
agents, insecticide, herbicides, and antibiotics (Crozier et al., 2008). In
(0.55 mg TE/g extract) as compared to methanol extract (0.24 mg TE/g
the current research work, the methanol and DCM extracts of S. fruticosa
extract). Furthermore, the DCM extract also presented superior metal
were tested for the determination of total phenolic and total flavonoid
chelating power with a value of 18.15 mg EDTAE/g extract.
contents, and the results are presented in Table 1. The higher amount of
phenolic (11.09 mg GAE/g extract) and flavonoid (20.50 mg QE/g
3.4. Enzyme inhibition studies
extract) contents were observed in the methanol extracts, while low
phenolic (9.25 mg GAE/g extract) and flavonoid (4.98 mg QE/g extract)
Alzheimer’s disease (AD) falls in the category of neurodegenerative
contents were observed in the case of DCM extracts (Table 1). A similar
disease, which is the main reason of gradual and irreversible damage to
type of results was reported in a previous study in which the higher
the brain (Parihar and Hemnani, 2004). AD is linked to aging and is
flavonoid contents were noted in the methanol extract (29.8 mg CE/g) of
accountable for damaging different parts of brain. It affects the cholin­
Reaumuria vermiculata as compared to DCM extracts (25.12 mg CE/g)
ergic neurons, and as result neurotransmitters transmission is stopped
(Karker et al., 2016).
(Querfurth and LaFerla, 2010; Ritter, 2012). AChE is in charge of con­
verting acetylcholine into choline and acetic acid. As a result, the
Table 1
enzyme AChE plays an important role in the development of AD. By
Total phenolic and flavonoid contents in Suaeda fruticosa extracts.
limiting acetylcholine breakdown, inhibitors stimulate neurotransmis­
Extracts Total phenolic content (mg GAE/g) Total flavonoid content (mg QE/g) sion (García-Ayllón et al., 2011). Butyrylcholinesterase is the second
SF-M 11.09 ± 0.08 20.50 ± 0.68 member of the cholinesterase family, but its functions have not been
SF-D 9.25 ± 0.07 4.98 ± 0.16 thoroughly investigated, and is found to be associated with the AD
SF-M: Suaeda fruticosa methanol extract; SF-D: Suaeda fruticosa DCM extract. (Karlsson, 2013). In the present study, the methanol and DCM extracts of
Data from three repetitions, with mean ± standard deviation; GAE: gallic acid S. fruticosa were tested for the AChE and BChE inhibition, and as pre­
equivalent; QE: quercetin equivalent. sented in Table 4, a considerable inhibition potential was observed for

3
H. Saleem et al. Food and Chemical Toxicology 154 (2021) 112348

Fig. 1. Total ion chromatogram (TIC) of Suaeda fruticosa methanol extract.

Table 2
UHPLC-MS of S. fruticosa methanol extract.
S.no RT (min) B. peak (m/z) Tentative compound identification Comp. class MFG formula Mol. mass

1 2.485 332.92 Costatol Alcohol derivative C10H14BrCl3O 333.92


2 2.729 135.03 D-threonic acid Carbohydrate C4H8O5 136.03
3 2.799 157.04 Allantoin Urea derivative C4H6N4O3 158.04
4 12.27 153.02 3,4-Dihydroxybenzoic acid Phenol C7H6O4 154.02
5 12.783 328.12 3-O-Acetylhamayne Alkaloid C18H19NO5 329.12
6 15.153 329.24 5,8,12-trihydroxy-9-octadecenoic acid Fatty acid C18H34O5 330.24
7 15.97 881.49 Pitheduloside B Saponin C46H74O16 882.49
8 16.67 267.13 Kamahine C Spiroketal C14H20O5 268.13
9 17.711 293.18 Gingerol Phenol C17H26O4 294.18
10 17.713 356.17 Uplandicine Alkaloid C17H27NO7 357.17
11 21.065 233.16 Curcumenol Guaianes C15H22O2 234.16

RT: retention time; B. peak: base peak.

Table 3
Antioxidant properties of Suaeda fruticosa extracts.
Extracts Radical Scavenging activity Reducing power Total antioxidant capacity (TAC) Ferrous chelating

DPPH (mgTE/g ABTS (mgTE/g FRAP (mgTE/g CUPRAC (mgTE/g Phosphomolybdenum (mgTE/g Metal Chelating
extract) extract) extract)) extract) extract) (mgEDTA/g)

SF-M 9.06 ± 0.63 16.46 ± 1.59 25.01 ± 0.49 48.89 ± 1.63 0.24 ± 0.02 1.77 ± 0.19
SF-D 5.69 ± 0.15 10.52 ± 0.20 23.84 ± 0.77 52.52 ± 1.23 0.55 ± 0.09 18.15 ± 1.18

TE: trolox equivalent; EDTAE: EDTA equivalent. Values expressed are means ± S.D. of three parallel measurements.

in the treatment of hyperpigmentation in humans, such as nevus, len­


Table 4 tigo, and post-inflammatory phases (Parvez et al., 2006). The methanol
Enzyme inhibition effects of Suaeda fruticosa extracts.
and DCM extracts of S. fruticosa were screened for tyrosinase inhibition,
Extracts AChE BChE Tyrosinase Glucosidase Amylase and the results are presented in Table 4. Both the extracts revealed
inhibition inhibition (mg KAE/g (mmol (mmol
comparatively same levels of inhibition with values of 125.38 mg KAE/g
(mg (mg extract) ACAE/g ACAE/g
GALAE/g GALAE/g extract) extract) extract for methanol extract and 123.12 mg KAE/g extract for the DCM
extract) extract) extract.
SF-M 4.70 ± 4.64 ± 125.38 ± 0.54 ± 0.02 1.94 ±
Glucosidases (including α-glucosidase and α-amylase enzymes) are
0.22 0.26 1.25 0.01 found in the gastrointestinal (GI) tract, where these enzymes are
SF-D 4.23 ± 4.73 ± 123.12 ± 0.61 ± 0.02 1.96 ± involved in catalyzing the final step of the digestion of carbohydrates.
0.05 0.09 0.63 0.01 Thus, in order to develop the compounds having inhibition potential
GALAE: galatamine equivalent; KAE: kojic acid equivalent; ACAE: acarbose against these enzymes is a promising approach (Mollica et al., 2019).
equivalent; all values expressed are means ± S.D. of three parallel Higher sugar levels in the blood can develop to diabetes, which is the
measurements. major cause of kidney, eye, and cardiovascular diseases. As a result, in
order to cure diabetes, blood sugar levels must be reduced. The search
both the methanol (AChE: 4.70 mg GALAE/g extract; BChE: 4.64 ± 0.26 for α-amylase and α-glucosidase inhibitors is of great significance in the
mg GALAE/g) and DCM (AChE: 4.23 mg GALAE/g extract; BChE: 4.73 treatment of diabetes (Bhandurge et al., 2010). In this research work, the
mg GALAE/g extract) extracts. methanol and DCM extracts of S. fruticosa were examined for α-gluco­
Tyrosinase is a copper-containing enzyme that is important for sidase, and α-amylase inhibition, and the results are depicted in Table 4.
catalyzing oxidation during the early stages of melanin production For α-glucosidase inhibition, a stronger level of inhibition potential was
(Parvez et al., 2006). Because of the prevalence of skin problems, the observed in DCM extract (0.61 mmol ACAE/g extract) as compared to
quest for tyrosinase inhibitors is critical. Tyrosinase inhibitors are useful methanol extract (0.54 mmol ACAE/g extract). Likewise, for the

4
H. Saleem et al. Food and Chemical Toxicology 154 (2021) 112348

α-amylase inhibition activity, almost similar values of inhibition were Table 5


noted for both the methanol (1.94 mmol ACAE/g extract) and DCM Cytotoxicity of Suaeda fruticosa methanol and DCM extracts against tested cell
(1.96 mmol ACAE/g extract) extracts. lines.
Samples % Viability (200 μg/mL)
3.5. Cytotoxicity MCF-7 MDA-MB-231 DU-145

SF-M 63.44 77.75 62.83


The cytotoxicity of both the methanol and DCM extracts of
SF-D 45.01 67.22 25.88
S. fruticosa was also evaluated against MCF-7 and MDA-MB-231 breast
cancer cell lines, and the results of the toxicity assay are presented in SF-M: Suaeda fruticosa methanol extract; SF-D: Suaeda fruticosa DCM extract;
Table 5. From the results, it is clear that all the tested extracts presented all values expressed are means ± S.D. of three parallel measurements.
low to moderate toxicity against the tested cell lines. The methanol
extract was noted to be the most cytotoxic against the tested MCF-7,
Table 6
MDA-MB-231, and DU-145 cell lines with percentage viability of Binding energy (kcal/mol), Inhibition constant Ki, interaction sites of the studied
63.44, 77.75, and 62.83%, respectively. Likewise, the DCM extract was compounds with AChE, BChE, tyrosinase, α-amylase and α-glucosidase enzymes.
noted to be most active for the MDA-MB-231 cell line. This is just a
Binding energy/ Interaction site
preliminary toxicity testing of the studied plant extract; the detailed in- inhibition constant Ki
vivo toxicity studies are recommended.
Uplandicine
AChE − 5.58 (81.63 μM) Ser 125(HB), Asn 87(HB), Tyr 337(HB),
3.6. Docking results Asp 74, Tyr 124, Phe 297, Phe 338, Gly
122(HB), Ser 203
Currently, docking is one of the efficient and reliable tools to predict BChE − 4.63 (405.60 μM) Tyr 332, Pro 85(HB), Gly 117(HB), His
binding free energy and to correlate the experimental biological activity. 438, Trp 82
Tyrosinase − 2.80 (8.94 mM) His 208, Val 218, Met 61, His 204, His 60
In this study, three selected compounds as the dominant compounds α-amylase − 2.88 (7.75 mM) Trp 83(HB), Arg 344(HB), Tyr 82, Leu 166,
were docked against AChE, BChE, tyrosinase, α-amylase, and α-gluco­ His 122, Asp206
sidase enzymes. The active site of these enzymes was detected and α-glucosidase − 4.34 (654.70 μM) Ala 234, Trp 329, Phe 601, Ala 602, Asp
confirmed by docking the control ligands. The docking results are shown 568(HB), Arg 552(HB) Asp 469(HB)
3,4-Dihydroxybenzoic acid
in Table 6 and Fig. 2. In Table 6, the calculated binding affinities of the
AChE − 4.54 (470.14 μM) His 447(HB), Tyr 337, Trp 86, Glu 202
studied compounds against the five enzymes are listed with the esti­ (HB)
mated inhibition constants. In addition, the residual amino acids BChE − 4.38 (616.55 μM) Thr 120(HB), Trp 82(HB), Tyr 128(HB),
involved in the interactions with the inhibitor at the active site are Glu 197(HB)
shown in Table 6. Uplandicine has shown the highest binding energy Tyrosinase − 4.36 (635.81 μM) His 208, His 69(HB), His 231(HB), Ala
221, Val 218
with the AChE enzyme (− 5.58 kcal/mol) and the lowest inhibition α-amylase − 4.41 (585.94 μM) Asp 340(HB), Asp 206(HB), Arg 204(HB),
constant (81.63 μM) in comparison with the rest of the studied enzymes. Tyr 82
This inhibition is mainly attributed to the high number of Hydrogen α-glucosidase − 4.93 (242.02 μM) Asp 357(HB), Asp 469, Asp 568(HB)
bonds formed with Ser 125, Asn 87, Tyr 337, and Gly 122 at the active 3-O-Acetylhamayne
AChE − 9.38 (133.91 nM) Tyr 124(HB), Tyr 341, Tyr 337, His 447,
site of the enzyme, as shown in Fig. 2. In contrast, 3,4-dihydroxybenzoic
Glu 202, Trp 86, Gly 120, Leu 130, Tyr 133
acid has shown similar affinity with the five enzymes as shown in (HB)
Table 6; however, the highest binding free energy is with α-glucosidase BChE − 8.84 (329.84 nM) Thr 122(HB), His 438, Ala 328, Trp 82
enzyme (− 4.93 kcal/mol) and inhibition constant (242.02 μM). Tyrosinase − 7.13 (5.96 μM) Asn 205 (HB), Phe 197, Arg 209, Gly 216,
Hydrogen bonds and pi-pi interactions are the dominant interactions Val 217, Val 218, His 208, His 60, His 204,
Ala 221
between this inhibitor and the active site of the studied enzymes. Similar α-amylase − 6.37 (21.52 μM) Asp 206(HB), Leu 173, Leu 166, Glu 230,
to uplandicine, 3-O-acetylhamayne has shown higher binding energy Leu 232, Asp 297
with AChE enzyme with binding free energy equal to (− 9.38 kcal/mol) α-glucosidase − 6.36 (21.87 μM) Asp 568(HB), Trp 432, Phe 476, Trp 329,
and inhibition constant (133.91 nM). The interactions with the active Phe 601
site involve hydrogen bonds with Tyr 124 and Tyr 133 residues. (HB)* = Hydrogen bond.

4. Conclusion Funding

In this study, the phytochemical, biological, and toxicological pa­ This research work is partly supported by the project number
rameters of methanol and DCM extracts S. fruticosa were explored. The (TURSP-2020/288), Taif University, Taif, Saudi Arabia.
methanol extract contains a considerable amount of total phenolic and
flavonoid contents and also presented considerable antioxidant poten­ CRediT authorship contribution statement
tial. All the extracts presented strong to moderate enzyme inhibition
potential against the tested enzymes. The UHPLC-MS secondary Hammad Saleem: Conceptualization, Methodology, Formal anal­
metabolite profiling of methanol extract revealed the tentative identi­ ysis, Investigation, Writing – original draft. Umair Khurshid: Writing –
fication of several important secondary metabolites. Both the tested original draft. Muhammad Sarfraz: Writing – review & editing.
extracts were found to be moderately cytotoxic against breast cancer cell Muhammad Imran Tousif: Writing – original draft. Abdulwahab
lines and prostate cancer cells. Theoretically, docking study has Alamri: Funding acquisition. Sirajudheen Anwar: Funding acquisition.
confirmed the inhibition ability of the three selected compounds with Abdulhakeem Alamri: Funding acquisition. Irshad Ahmad: Funding
the AChE, BChE, tyrosinase, α-amylase, and α-glucosidase enzymes and acquisition. Hassan H. Abdallah: Formal analysis, Investigation. Fawzi
their interactions with the active site are elucidated. Further in­ M. Mahomoodally: Formal analysis. Nafees Ahemad: Conceptualiza­
vestigations can be done for the isolation of new potentially bioactive tion, Supervision, Funding acquisition.
compounds.

5
H. Saleem et al. Food and Chemical Toxicology 154 (2021) 112348

Fig. 2. The docked compounds at the active site of the enzyme and their interactions.

Declaration of competing interest Karker, M., Falleh, H., Msaada, K., Smaoui, A., Abdelly, C., Legault, J., Ksouri, R., 2016.
Antioxidant, anti-inflammatory and anticancer activities of the medicinal halophyte
Reaumuria vermiculata. EXCLI journal 15, 297.
I Hammad Saleem is submitting a manuscript entitled for possible Karlsson, D., 2013. Biomolecular Screening for Inhibitors of Butyrylcholinesterase:
publication in Food and Chemical Toxicology. It is stated that there is no Identification and Characterization Using in Vitro and in Silico Tools.
Conflict of Interest for the submitted paper. Ksouri, R., Falleh, H., Megdiche, W., Trabelsi, N., Mhamdi, B., Chaieb, K., Bakrouf, A.,
Magné, C., Abdelly, C., 2009. Antioxidant and antimicrobial activities of the edible
medicinal halophyte Tamarix gallica L. and related polyphenolic constituents. Food
References Chem. Toxicol. 47, 2083–2091.
Mollica, A., Zengin, G., Durdagi, S., Ekhteiari Salmas, R., Macedonio, G., Stefanucci, A.,
Abirami, A., Nagarani, G., Siddhuraju, P., 2014. In vitro antioxidant, anti-diabetic, Dimmito, M.P., Novellino, E., 2019. Combinatorial peptide library screening for
cholinesterase and tyrosinase inhibitory potential of fresh juice from Citrus hystrix discovery of diverse α-glucosidase inhibitors using molecular dynamics simulations
and C. maxima fruits. Food Science and Human Wellness 3, 16–25. and binary QSAR models. J. Biomol. Struct. Dyn. 37, 726–740.
Ahmad, R., Malik, K.A., 2013. Prospects for Saline Agriculture. Springer Science & Mollica, A., Zengin, G., Locatelli, M., Stefanucci, A., Mocan, A., Macedonio, G.,
Business Media. Carradori, S., Onaolapo, O., Onaolapo, A., Adegoke, J., 2017. Anti-diabetic and anti-
Alho, H., Leinonen, J., 1999. Total antioxidant activity measured by chemiluminescence hyperlipidemic properties of Capparis spinosa L.: in vivo and in vitro evaluation of its
methods. Methods in Enzymology. Elsevier, pp. 3–15. nutraceutical potential. Journal of Functional Foods 35, 32–42.
Ashraf, M.T., Fang, C., Bochenski, T., Cybulska, I., Alassali, A., Sowunmi, A., Naija, D.S., Bouzidi, A., Boussaada, O., Helal, A.N., Mahjoub, M.A., Echafai, N.,
Farzanah, R., Brudecki, G.P., Chaturvedi, T., Haris, S., 2016. Estimation of bioenergy Mighri, Z., 2014. The antioxidant and free-radical scavenging activities of Tamarix
potential for local biomass in the United Arab Emirates. Emir. J. Food Agric. 99–106. boveana and Suaeda fruticosa fractions and related active compound. European
Bareen, F.-e., Tahira, S.A., 2011. Metal accumulation potential of wild plants in tannery Scientific Journal, ESJ 10.
effluent contaminated soil of Kasur, Pakistan: field trials for toxic metal cleanup Nemudzivhadi, V., Masoko, P., 2014. In vitro assessment of cytotoxicity, antioxidant, and
using Suaeda fruticosa. J. Hazard Mater. 186, 443–450. anti-inflammatory activities of Ricinus communis (Euphorbiaceae) leaf extracts. Evid.
Bennani-Kabchi, N., Kehel, L., Fdhil, H., Marquie, G., 1999. Effect of Suaeda fruticosa base Compl. Alternative Med. 2014.
aqueous extract in the hypercholesterolaemic and insulin-resistant sand rat. Parihar, M., Hemnani, T., 2004. Alzheimer’s disease pathogenesis and therapeutic
Therapie 54, 725–730. interventions. J. Clin. Neurosci. 11, 456–467.
Bhandurge, P., Rajarajeshwari, N., Alagawadi, K., Agrawal, S., 2010. Antidiabetic and Parvez, S., Kang, M., Chung, H.S., Cho, C., Hong, M.C., Shin, M.K., Bae, H., 2006. Survey
hyperlipaemic effects of Citrus Maxima Linn fruits on alloxan-induced diabetic rats. and mechanism of skin depigmenting and lightning agents. Phytother Res.: An
Int. J. Drug Dev. Res. 2, 273–278. International Journal Devoted to Pharmacological and Toxicological Evaluation of
Crozier, A., Clifford, M.N., Ashihara, H., 2008. Plant Secondary Metabolites: Occurrence, Natural Product Derivatives 20, 921–934.
Structure and Role in the Human Diet. John Wiley & Sons. Qasim, M., Gulzar, S., Khan, M.A., 2011. Halophytes as Medicinal Plants. NAM Meeting
Duh, P.-D., 1998. Antioxidant activity of burdock (Arctium lappa Linne): its scavenging in Denizli, Turkey.
effect on free-radical and active oxygen. J. Am. Oil Chem. Soc. 75, 455–461. Querfurth, H.W., LaFerla, F.M., 2010. Mechanisms of disease. N. Engl. J. Med. 362,
García-Ayllón, M.-S., Small, D.H., Avila, J., Sáez-Valero, J., 2011. Revisiting the role of 329–344.
acetylcholinesterase in Alzheimer’s disease: cross-talk with P-tau and β-amyloid. Ritter, J.M., 2012. Drugs for Alzheimer’s disease. Br. J. Clin. Pharmacol. 73, 501–503.
Front. Mol. Neurosci. 4. Saleem, H., Zengin, G., Locatelli, M., Mollica, A., Ahmad, I., Mahomoodally, F.M.,
Goodall, D.W., Perry, R.A., Howes, K.M.W., 2009. Arid Land Ecosystems: Volume 2, Abidin, S.A.Z., Ahemad, N., 2019. In vitro biological propensities and chemical
Structure, Functioning and Management. Cambridge University Press. profiling of Euphorbia milii Des Moul (Euphorbiaceae): a novel source for bioactive
Grata, E., Guillarme, D., Glauser, G., Boccard, J., Carrupt, P.-A., Veuthey, J.-L., Rudaz, S., agents. Ind. Crop. Prod. 130, 9–15.
Wolfender, J.-L., 2009. Metabolite profiling of plant extracts by ultra-high-pressure van der Watt, E., Pretorius, J.C., 2001. Purification and identification of active
liquid chromatography at elevated temperature coupled to time-of-flight mass antibacterial components in Carpobrotus edulis L. J. Ethnopharmacol. 76, 87–91.
spectrometry. J. Chromatogr. A 1216, 5660–5668. Weber, D., Ansari, R., Gul, B., Khan, M.A., 2007. Potential of halophytes as source of
Grochowski, D.M., Uysal, S., Aktumsek, A., Granica, S., Zengin, G., Ceylan, R., edible oil. J. Arid Environ. 68, 315–321.
Locatelli, M., Tomczyk, M., 2017. In vitro enzyme inhibitory properties, antioxidant Wickens, G.E., Field, D.V., Goodin, J.R., 2012. Plants for Arid Lands: Proceedings of the
activities, and phytochemical profile of Potentilla thuringiaca. Phytochemistry Letters Kew International Conference on Economic Plants for Arid Lands Held in the Jodrell
20, 365–372. Laboratory, Royal Botanic Gardens, Kew, England, 23–27 July 1984. Springer
Halliwell, B., 1990. How to characterize a biological antioxidant. Free Radic. Res. Science & Business Media.
Commun. 9, 1–32. Yadav, R., Agarwala, M., 2011. Phytochemical analysis of some medicinal plants.
Halliwell, B., 1995. Antioxidant characterization: methodology and mechanism. J. Phytol.
Biochem. Pharmacol. 49, 1341–1348. Zengin, G., Atasagun, B., Aumeeruddy, M.Z., Saleem, H., Mollica, A., Bahadori, M.B.,
Hameed, A., Hussain, T., Gulzar, S., Aziz, I., Gul, B., Khan, M.A., 2012. Salt tolerance of a Mahomoodally, M.F., 2019. Phenolic profiling and in vitro biological properties of
cash crop halophyte Suaeda fruticosa: biochemical responses to salt and exogenous two Lamiaceae species (Salvia modesta and Thymus argaeus): a comprehensive
chemical treatments. Acta Physiol. Plant. 34, 2331–2340. evaluation. Ind. Crop. Prod. 128, 308–314.
Irwin, J.J., Sterling, T., Mysinger, M.M., Bolstad, E.S., Coleman, R.G., 2012. ZINC: a free Zengin, G., Nithiyanantham, S., Locatelli, M., Ceylan, R., Uysal, S., Aktumsek, A.,
tool to discover chemistry for biology. J. Chem. Inf. Model. 52, 1757–1768. Selvi, P.K., Maskovic, P., 2016. Screening of in vitro antioxidant and enzyme
Jalas, J., Suominen, J., 1988. Atlas Florae Europaeae: Volume 3: Distribution of Vascular inhibitory activities of different extracts from two uninvestigated wild plants:
Plants in Europe. Cambridge University Press. Centranthus longiflorus subsp. longiflorus and Cerinthe minor subsp. auriculata.
European Journal of Integrative Medicine 8, 286–292.

You might also like